Skip to main content
Journal of Cellular and Molecular Medicine logoLink to Journal of Cellular and Molecular Medicine
. 2009 Dec 8;15(8):1645–1687. doi: 10.1111/j.1582-4934.2009.00987.x

Growth factors in ischemic stroke

S Lanfranconi a,*, F Locatelli b, S Corti a, L Candelise a, G P Comi a, P L Baron a, S Strazzer b, N Bresolin a,b, A Bersano a,*
PMCID: PMC4373358  PMID: 20015202

Abstract

Data from pre-clinical and clinical studies provide evidence that colony-stimulating factors (CSFs) and other growth factors (GFs) can improve stroke outcome by reducing stroke damage through their anti-apoptotic and anti-inflammatory effects, and by promoting angiogenesis and neurogenesis. This review provides a critical and up-to-date literature review on CSF use in stroke. We searched for experimental and clinical studies on haemopoietic GFs such as granulocyte CSF, erythropoietin, granulocyte-macrophage colony-stimulating factor, stem cell factor (SCF), vascular endothelial GF, stromal cell-derived factor-1α and SCF in ischemic stroke. We also considered studies on insulin-like growth factor-1 and neurotrophins. Despite promising results from animal models, the lack of data in human beings hampers efficacy assessments of GFs on stroke outcome. We provide a comprehensive and critical view of the present knowledge about GFs and stroke, and an overview of ongoing and future prospects.

Keywords: stroke, growth factors, G-CSF, EPO, GM-CSF, SCF, VEGF, IGF-1, SDF-1α

Background

Stroke is a leading cause of death and disability worldwide among the adult population. Despite the recognised efficacy of some interventions in the acute phase, the narrow therapeutic window makes these treatments applicable to only a minority of stroke patients. Once the infarct is established, no therapy can help recover or replace the lost tissue, and rehabilitation and pharmacological treatment of co-morbidities are the only therapeutic strategies remaining for most stroke patients.

Thus, new approaches to restore function after stroke are under investigation. Some evidence from pre-clinical studies suggests that stem cell transplantation may represent a potential option for stroke treatment. However, several limitations, including experimental conditions for the in vitro culture of neural stem cells, choice of stem cell type, best route of administration and establishment of neuronal connections make it difficult to apply this treatment in a clinical setting [1, 2]. Moreover, since cell transplantation also requires surgical intervention in some cases, it is desirable to explore less invasive therapeutic strategies. The results of the first pre-clinical and clinical studies on stem cell transplantation highlight that cell differentiation, survival and trophic cell support is promoted by growth factors (GFs). In endogenous neurogenesis, GFs induced proliferation and differentiation of adult neural stem cells from the sub-ventricular zone of lateral ventricles (SVZ) and the dentate gyrus (DG) of the hippocampus into mature neurons in both animal models [36] and human studies [7, 8]. These findings suggest that GF therapy could represent an alternative therapeutic approach to promote migration and differentiation and to enhance the survival of endogenous stem cells by modulating pathways of endogenous neurogenesis.

Experimental studies show that hematopoietic GFs can improve stroke outcome through their pleiotropic effects which include neuroprotection, stem cell survival and promotion of angiogenesis and neurogenesis, as well as through their anti-apoptotic and anti-inflammatory effects [9].

Most GFs act binding specific receptors activating different signalling pathways and inducing the expression of specific genes involved in cell proliferation and differentiation. For instance, receptor-mediated activation of the mitogen-activated protein kinase (MAPK) promotes proliferation [10], whereas stimulation of the phosphoinosotide-dependent kinase/Akt pathway induces differentiation of stem cells. Thus, the response to specific GFs is influenced by the expression of their corresponding receptors on target cells [11, 12].

This review provides a critical, up-to-date evaluation of the literature relevant to the role of select GFs in post-stroke recovery. Both experimental models and human studies of ischemic stroke are included and discussed.

Search strategies

We included studies (abstracts, letters, articles, case–control studies, reviews and meta-analyses) on experimental models of stroke and in human beings. Our search focused on the GFs most often studied in ischemic stroke. The literature search included articles from 1960 to October 2009 in electronic bibliographic databases (MEDLINE, EMBASE). Reference lists from primary and review articles, and the MEDLINE function ‘related articles’ were also consulted. As search key words, we used: granulocyte colony-stimulating factor (G-CSF), erythropoietin (EPO), granulocyte-macrophage colony-stimulating factor (GM-CSF), stem cell factor (SCF), vascular endothelial growth factor (VEGF), stromal cell-derived factor-1α (SDF-1α) and insulin growth factor-1 (IGF-1), and ischemic stroke, stroke, cerebrovascular disease and stem cell transplantation. Finally, we also examined non-English articles and studies on haemorrhagic stroke.

Haematopoietic growth factors

Red cells, granulocytes, monocytes, platelets and lymphocytes all derive from a common multipotent bone-marrow stem cell. Haemopoietic GFs, also known as CSFs, modulate lineage-specific differentiation of bone marrow stem cells (BMSCs), leading to the generation of circulating red cells, white cells and platelets.

Data from experimental studies (Table 1) support the notion that CSFs could improve stroke outcome by reducing stroke damage and improving post-stroke brain repair [9].

Table 1.

Growth factors experimental studies

Growth factors Study reference Animal model Sample size (treatment group/control group) Type of study (focal or global ischemia) Treatment Time of administration Results
G-CSF [51] Rat 12/12 90-min. MCAO GCS-F 60 mcg/kg i.v. within 24 hrs 30 min. after the induction of ischemia 47% reduction of infarct volume (132.0 ± 112.7 mm3 in the G-CSF group versus 278.9 ± 91.6 mm3 in the vehicle group). Up-regulation of STAT3 in the peri-ischemic area.
[52] Mouse 12/15 60-min. MCAO GCS-F 50 mcg/kg s.c. within 24 hrs or vehicle 24 hrs after occlusion Reduction of infarct size (27 ± 7 mm3; n = 9 in the G-CSF group versus 69 ± 5 mm3; n = 3 in the vehicle group). Significant increase in survival rate (75% versus 20% in the treatment group).
[68] Rat 15/15 60-min. MCAO GCS-F 50 mcg/kg s.c. within 24 hrs or vehicle 24 hrs after occlusion Infarct volume reduction (61 ± 12 mm3 in G-CSF–treated animals versus 176 ± 20 mm3 in the vehicle group). Significantly increased numbers of BrdU+ cells in their ipsilateral hemispheres in the treatment versus control group. Improved neurological behavior.
[53] Mice 12/15+6 (sham-operated) 60-min. MCAO G-CSF injected s.c. (50 mcg/kg) or vehicle 1 hr after MCAO 46% reduction of infarct size (14.91 ± 3.5 mm3 in the G-CSF group versus 27.66 ± 8.79 mm3 in the vehicle group). Significant improvement in motor task.
[63] Rat 139/67 90-min. MCAO G-CSF 50 mcg/kg i.p. in first 24 hrs (total of 150 mcg/kg) 2, 24, 96 or 168 hrs after occlusion Reduction in infarct volume (106.9 ± 59.6 mm3 in the treatment group versus controls 222.9 ± 80.4 mm3), in hemispheric atrophy at 35 days and in BBB damage. Increased fraction of BrdU+ endothelial cells. Significant increase in endothelial cells proliferation and in mean diameter of the cerebral microvessels in the ipsilateral hemisphere. eNOS and Ang2 up-regulation, Improved functional recovery.
[36] Rat 15/15 90-min. MCAO Direct distal MCAO Photothrombotic induction of ischemia G-CSF 60 mcg/kg i.v. within 24 hrs G-CSF 60 mcg/kg i.v. within 24 hrs G-CSF 15 mcg/kg i.v. for 5 days 2 hrs after occlusion 1 hr after occlusion Infarct size reduction (15% versus 37%). Increased recruitment of neural progenitors. Improved functional outcome after cerebral ischemia.
[38] Mice 35/35 60-min. MCAO Recombinant human G-CSF (50 mcg/kg) intravascularly or vehicle 30 min. after vessel occlusion Infarct size reduction (18.84 ± 3.72 mm3 in the treatment group versus 28.59 ± 3.26 mm3 in the control group). Increase in a time-dependent manner STAT3 expression. Stronger Bcl-2 and iNOS expression in the transition area in the G-CSF group.
[54] Rat 90-min. MCAO Photothrombotic induction of ischemia in the sensorimotor cortex. GCS-F 60 mcg/kg i.v. within 24 hrs G-CSF 10 mcg/kg i.v 4 hrs after occlusion 1, 24 and 72 hrs after ischemia for 10 days. 33% infarct size reduction (223.33 ± 27.3 mm3 in the G-CSF group versus 334.0 ± 31.5 mm3 in the vehicle group). Improvement in sensorimotor deficits at the rotarod test.
[39] Rat 55 90-min. MCAO Two groups: single dose of G-CSF s.c. (50 mcg/kg) or vehicle; G-CSF s.c. (50 mcg/kg) or vehicle for two additional days At the onset of reperfusion or at the onset of reperfusion and for additional 3 days Reduction of early neurological deficits, attenuation of infarct volume, increased neuronal and glial survival by activating different anti-apoptotic ways
[55] Rat 56/40 60-min. MCAO G-CSF 10 mcg/kg s.c. within 24 hrs (total 50 mcg/kg) 6 hrs after occlusion Non-significant infarct size reduction. Significant increase of the survival rate and of the neurological improvement. Increased number of CD34+ cells in the marginal zone of the infarction at 7, 14 and 21 days after cerebral ischemia.
[64] Rat 5/5 90-min. MCAO Single dose G-CSF injected s.c. (50 mcg/kg) or vehicle With reperfusion Increased number of BrdU+ cells in the G-CSF-treated group.
[65] Rat 10/10 90-min. MCAO G-CSF injected s.c. (50 mcg/kg) or vehicle After reperfusion 31% reduction infarct volume 31% (25.1 ± 12.1% in the G-CSF group versus 56.5 ± 13.7% in the control group). Increased BrdU+ cells at 28 days in the G-CSF group. Increased NeuN plus BrdU double-positive cells and in the G-CSF-group at 7 days. Increase in BrdU+ endothelial cells in G-CSF-treated group (16.0 ± 4.5/mm2) compared with vehicle-treated group (7.0 ± 2.0/mm2), no longer persisting at 28 days.
[60] Rat 20/20+5 90-min. MCAO G-CSF injected s.c. (50 mcg/kg) or vehicle Immediately after riperfusion 55.6% reduction in infarct volume 119.4 ± 44.9 mm3 in the G-CSF group versus 269.1 ± 42.1 mm3 in the control group. G-CSF treatment improved the neurologic outcome by 50% at 24 hr. Significant reduction of TNF-a-positive cells at 8 to 72 hr and TGF-b2- and iNOS-positive cells at 24 and 72 hr after MCAO in the peri-ischemic area.
[62] CB-17 mice MCAO Human recombinant G-CSF s.c. (0.5, 5, 50 or 250 mcg/kg)/continuous administration (100 mcg/kg/day) by micro-osmotic pump or PBS or recombinant human EPO At 24, 48 and 72 hrs after stroke or 1 hr after stroke (at doses of 0.5, 5, 50 or 250 mcg/kg) or 1 hr after stroke over 7 days Negative effect on brain injury, brain atrophy and on functional recovery after stroke, enhancement of inflammatory response, positive effect on angiogenesis.
[61] BL/6J mice 13+11/33 MCAO Recombinant human G-CSF 300 mcg/kg and recombinant murine SCF 100 mcg/kg or vehicle Acute phase (days 1–10) and in the subacute phase (days 11–20 after the occlusion) Increased expression of mRNA IL-10 and anti-inflammatory cytokines.
[66] Mice Permanent MCAO after sex-mismatched bone marrow transplantation from EGFP-expressing mice G-CSF/SCF treatment G-CSF/SCF treatment reduced infarct volumes by more than 50%, resulted in a 1.5-fold increase in vessel formation, led to a 2-fold increase in the number of newborn cells in the ischemic hemisphere.
[35] Male Wistar rats 15 60 min. temporary MCAO i.v. saline or G-CSF (60 mcg/kg) 30 min. after temporary MCAO G-CSF significantly attenuated the release of glutamate in the infarcted striatum from 30 min. to 180 min. after tMCAO compared with controls (P < 0.05). Infarct volume reduced significantly compared to controls at 24 hr after tMCAO.
EPO [110] Rat 60 min. MCAO i.p. administration of EPO (5,000 units/kg of body weight, i.p.) At the time of occlusion Reduction of the volume of infarction, almost complete reduction in the number of terminal deoxynucleotidyltransferase-mediated dUTP nick-end labeling of neurons within the ischemic penumbra.
[118] 7 days old Rat right CCAO i.p. DFO (200 mg/kg), recombinant human EPO (1 kU/kg), a combination of DFO-EPO or vehicle 0, 24 and 48 hrs after hypoxia-ischemia DFO-EPO administration reduced the number of cleaved caspase 3-positive cells in the ipsilateral cerebral cortex. However, treatment with DFO, EPO, or with the combination of DFO and EPO did not protect against grey or white matter damage in the experimental setting applied.
[120] Rat MCAO rhEPO; 5,000 U/kg body weight, i.p. at the time of MCA occlusion significant reduction of astrocytes, leucocytes and microglia activation as well as a decreased production of pro-inflammatory cytochines such as TNF-α, IL-6 and monocite chemoattractant protein 1 in ischemic area
[122] Rat Focal cerebral ischemia i.v. treatment with CEPO At different time points after MCA occlusion Reduced cellular infiltration, reduced apoptosis and white matter damage
VEGF [203] Rat 7/10 MCAO by placement of placement of a clot rhVEGF 1 mg/kg over a 4 hr interval (5 μl/min) or vehicle 1 hr or 48 hrs after embolization Enhancement of angiogenesis in the ischemic brain and reduction of neurological deficits during stroke recovery
[200] Rat 6/5 90-min. MCAO VEGF (1.0 ng/μl, 9 μl) was topically applied on the surface of the reperfused rat brain or vehicle After reperfusion Reduction in infarct volume (18.4% ± 2.1% in the VEGF group versus 33.2% ± 4.2% in the control group) and brain edema (81.5% ± 0.8% in the VEGF group versus 84.9% ± 2.1% in the vehicle treated group). Anti-apoptotic effect demonstrated by a significant decrease in TUNEL staining both at 24 (32.8 ± 8.5 cells/mm2 versus 46.4 ± 7.2 cells/mm2 in the control group) and 48 hr (40.7 ± 12.5 cells/mm2 versus 68.1 ± 13.6 cells/mm2 in the non-VEGF group).
[207] adult male CD-1 mice Six groups of 6 adult male CD-1 mice underwent 1) AdlacZ (viral vector control), 2) AdVEGF, 3) AdAng2, 4) VEGF protein, 5) VEGF protein plus AdAng2, 6) saline (negative control) injection 2 mcL of adenoviral suspension (AdVEGF, AdAng2, or AdlacZ) were injected stereotactically into the right caudate/putamen. For the protein infusion a i.c.v. infusion via osmotic mini-pump was used. Mice treated with VEGF protein infusion plus AdAng-2 significantly increased microvessel counts relative to all other groups (P < 0.05). Combination of VEGF and Ang-2 may lead to BBB disruption because it increases MMP-9 activity and inhibits ZO-1 expression.
[211] Adult C57/BL6 mice Animals were exposed to normobaric hypoxia at 12 ± 6% oxygen for 24 hrs or were kept at room air pressure A neutralizing goat antimouse VEGF antibody or a corresponding control antibody was injected i.p. at 100 mg Immediately prior to the 24 hrs hypoxic or normoxic exposure. Hypoxic exposure led to a significant increase in the levels of VEGF mRNA and protein in mouse brain that correlated with the severity of the hypoxic stimulus. Inhibition of VEGF activity by a neutralizing antibody completely blocked the hypoxia-induced increase in vascular permeability.
[212] Male Wistar rats 10 (VEGF antagonist treated)/15 (PBS treated) 2-VO model in which 2 adjacent cortical veins were photochemically occluded i.p. injection Immediately after 2-VO In treated animals there were attenuated vascular permeability and reduced cortical venous infarct in the acute stage.
[214] Gerbil model of focal brain ischemia 24+24 treated/24+24 controls A single 3-min. bilateral CCAO initially (about 4–5 min.) Animals were treated in the left lateral cerebral ventricle with rAAV-LacZ (control) or with rAAV-VEGF (0.5–25 U). Pre-treatment (6 days or 12 days) Improved survival, brain edema and delayed neuronal death in treated animals.
[205] Rat Group 1 (ischemia-only = 12), group 2 (ischemia + VEGF = 12), group 3 (ischemia + NSCs = 12), group 4 (ischemia + NSCs + VEGF = 12) Intraluminal thread occlusion of MCA for 90 min. i.v. administration of human neural stem cells (hNSCs: 5 × 106) and/or i.v. recombinant human VEGF [50 mcg/kg, 1 mcg/(kg min)]. hNSCs: 24 hrs after surgery VEGF: 48 hrs after surgery Better functional recovery, reduced cerebral atrophy and increased vascular density without a significant effect on stem cells survival, in hNSC+VEGF combined treated animals compared to the other groups.
[199] Rat 5+6+6/5+7+6 controls 2-hr MCAO Recombinant human VEGF 165 (5 mcg/ml), or vehicle was infused into the right lateral ventricle by osmotic minipump 1 week before MCAO I.c.v. infusion of VEGF 165 decreases infarct volume and brain edema after temporary MCAO without a significant increase in CBF.
[202] Mice 15/10 90-min. MCAO VEGF (8 ng i.c.v.) i.v. VEGF (15 ng/kg) 1 or 3 hrs after reperfusion Decreased infarct volume and improvement in neurological disability score. Enhancement of infarct volume.
[204] Rat 90-min. MCAO VEGF (10 μg/ml) was administered at 1 μl/hr i.c.v. For 3 days Improvement of both sensorimotor and cognitive functional outcome.
[216] Rat 3 animals per 10 groups temporary MCAO VEGF165 was administered intra-arterially for 7 days by miniosmotic pump: total dosages: 2 μg (low dose), 8 μg (intermediate dose), or 60 μg (high angiogenetic dose) After MCAO Non-angiogenic VEGF 165 treated brains showed preserved neuropil and reduced numbers of macrophages; ischemic brains treated by high angiogenic dose showed phagocytized neuropil and high macrophage density.
[198] Rat 144 30-min. MCAO i.c.v. injection of 5 mcg human VEGF 165-expressive plasmid (phVEGF) mixed with liposome At 15, 120, or 360 min. after ischemic operation VEGF overproduction improves stroke-induced striatal neurogenesis and enhances their maturation
[210] Rat 14/14 120-min. MCAO 3 × 106 MSCs in 1 ml total fluid volume PBS or vehicle 24 hrs after MCAO MSCs treatment promotes angiogenesis and vascular stabilization, which is at least partially mediated by VEGF/Flk1 and Ang1/Tie2.
[220] Rat 5+5/5 Transient MCAO Intracerebral administration of VEGF gene-transferred BMSCs engineered with a replication-deficient HSV type 1 1764/4-/pR19-VEGF 165 vector or native BMSCs, or vehicle 24 hrs after MCAO More potent autologous cell transplantation therapy than the transplantation of native BMSCs alone (significant functional recovery, lower infarct volume, stronger expression of VEGF proteins).
[201] Rat 90 min. of MCAO Pretreatment with KDR kinase inhibitor (Compound-1) (40 mg/kg p.o.) Starting 0.5 hr before occlusion Early pre-treatment administration of a KDR kinase inhibitor elicited an early, transient decrease in edema and subsequent reduction in infarct volume, implicating VEGF as a mediator of stroke-related vascular permeability and ischemic injury.
[218] VEGF(hi/+) and VEGF(lo/+) mice Femoral artery ligation MCA ligation Compared to VEGF(hi/+) mice, VEGF(lo/+) formed fewer collaterals during the perinatal period when adult density was established, and had 2-fold larger infarctions after MCA ligation, suggesting that VEGF and VEGFR-1 are determinants of arteriogenesis. VEGF-A specifies formation of native collaterals in healthy tissues.
[215] Wistar rats 93 MCAO for 2 hrs rAAV-VEGF, rAAV-null or physiologic saline was delivered into the lateral ventricle 8 weeks before MCAO I.c.v. rAAV-VEGF pre-treatment results in deleterious intracranial hypertension and augments secondary ischemic insults at the early stage of MCAO.
[219] Rat 4 for each condition NPCs expressing VEGF-A165 transplantation into the caudate putamen (1,000,000 cells) Increased survival of adenovirally transfected NPCs after 11 days, but not after 24 hr or 4 days. Increased expression of the endothelial cell marker PECAM-1 (CD31) after 24 hrs, 4 days, and 11 days after transplantation.
[213] Rat Stereotaxic injection of VEGF 1 mcg into the left SN VEGF administration highly up-regulated AQP4 mRNA and protein in the ventral midbrain, localized in close proximity to the VEGF-induced new blood vessels.
[217] Squirrel monkeys 3/4 Bipolar electrocoagulation of cortical vessels supplying the M1 hand area representation HIF-1a up-regulation is confined to the infarct and peri-infarct regions. Increases in VEGFR-2 immunoreactivity occurred in two remote regions.
GM-CSF [162] Rat 16/18 Bilateral vertebral and unilateral CCAO followed by hemodynamic stroke Daily injections of GM-CSF or vehicle Induction of arteriogenesis
[157] Rat Combined CCA/distal MCAO with 180 min. occlusion followed by 72 hrs reperfusion or proximal MCAO with 90 min. occlusion and 72 hrs reperfusion 10 mg/kg bodyweight GM-CSF i.v. over a time period of 20 min. or vehicle GM-CSF provides protection against experimental stroke and counteracts programmed cell death.
[161] Rat 13/8+8 Occlusion of vertebral plus left carotid artery GM-CSF or vehicle applied intraoperatively by a single intra-arterial dose into the non-occluded CA and subsequently by s.c injection every day GM-CSF treatment did not influence the arterial angioarchitecture on the dorsal brain surface but increased vessel diameter during natural arteriogenesis
[155] Rat 25/23 1 hr intraluminal MCAO Intracarotid injection of GM-CSF (5 ng) or saline Immediately after reperfusion Reduced the infarct volume and improved neurological function at 48 hrs after reperfusion in treated animals. Increased number of activated microglia/macrophages and decreased number of apoptotic cells in the penumbra area.
[163] Rat Not known BCAO BCAO and daily subcutaneous injection with GM-CSF 10 mcg/kg diluted in 0.3 ml saline, or vehicle, or sham occlusion and vehicle Injection started with occlusion of the first carotid artery until 5 weeks after the second CAO CBF and cerebrovascular reserve capacity recovered completely in GM-CSF–treated animals but not in solvent-treated animals. The capillary density showed a mild increase in GM-CSF–treated animals. The number of intraparenchymal and leptomeningeal arterioles was significantly higher in GM-CSF–treated animals than in both other groups.
[160] Rat model of post-natal hypoxia–ischemia Hypoxia–ischemia (HI, n = 67), hypoxia–ischemia with G-CSF treatment (HI+G, n = 65), healthy control (C, n = 53). Right CCAligation G-CSF (50 μg/kg s.c.) Start 1 hr after HI and given on 4 subsequent days Improved quantitative brain weight and qualitative Nissl histology. Decreased apoptotic cells (TUNEL positive), with reduced expression of Bax, cleaved caspase-3 and with increased expression of STAT3, Bcl-2, and Pim-1 in HI+G treated animals.
[158] Adult male C57BL/6 strain mice Unilateral CAAO Injection of GM-CSF (20 mcg/kg s.c.) or saline injection Every second day after CCAO until 7 days after CCA0 GM-CSF treatment produced increased leptomeningeal collateral growth, an increase in the number of Mac-2+ monocytes/macrophages on the surface of the brain and decreased infarct size
[159] unilateral MCAO GM-CSF at 60 mug/kg Daily for 5 consecutive days beginning immediately after injury GM-CSF leaded to decrease the extent of neuronal apoptosis by modulating the expression of several apoptosis-related genes such as Bcl-2, Bax, caspase 3, and p53, resulting in decreased infarction volume and improved locomotor behavior.
SCF [59] GFP-bone marrow engrafted mice (into the tail vein after irradiation) 6 for each treated group 6 for each control group Permanent MCAO 300 mcg/kg of recombinant human G-CSF and/or 100 mcg/kg of recombinant murine SCF. BrdU (50 mg/kg per day, s.c.) Cytokines: in the acute phase (days 1 to 10) or subacute phase (days 10 to 20 days) after MCAO BrdU: together with cytokines for 2 days, and 24 hrs after the last injection Subacute phase treated animals improved motor performance and higher brain function, compared with acute-phase treatment. Acute-phase and subacute-phase treatments identically reduced the infarct volume relative to vehicle treatment. Subacute-phase treatment significantly induced transition of bone marrow derived neuronal cells into the peri-infarct area and stimulated proliferation of intrinsic neural stem/progenitor cells in the neuroproliferative zone.
[260] Rat Saline, SCF, G-CSF, and SCF+G-CSF (n = 10/group) Permanent right MCA ligation Recombinant rat SCF (200 mcg/kg) and/or recombinant human G-CSF (50 mcg/kg) s.c. Start at 14 weeks after brain ischemia, for 7 days. Significant functional improvement was seen in SCF+G-CSF-treated rats 1, 5, and 17 weeks after injections. SCF alone also improved but not stable functional outcome. No functional benefit was seen in G-CSF-treated rats. Infarction volume was significantly reduced in SCF+G-CSF-treated rats.
[261] Rat 1° experiment: saline control, SCF, G-CSF, and SCF+G-CSF (n = 10), Sham-operative control rats (n = 6) 2° experiment: saline, SCF, G-CSF, or SCF+G-CSF (n = 4) Permanent right MCA ligation 1° experiment: s.c. injection of saline or GFs (recombinant rat SCF, 200 mcg/kg; recombinant human G-CSF, 50 mcg/kg). 2° experiment: s.c. injection of GFs plus BrdU; 50 mg/kg per day, i.p. 1°-2° experiments: 3 hrs to 7 days after brain ischemia. SCF-treated rats showed the best functional restoration at 1, 4, 7 and 10 weeks after the final injection. G-CSF-induced limited and unstable functional recovery. Stable but delayed functional improvement in SCF+G-CSF-treated rats. Infarction size was significantly reduced in all GF-treated rats. SCF and SCF+G-CSF enhanced NPC proliferation in the sub-ventricular zone bilaterally, whereas G-CSF and SCF+G-CSF treatment increased – BrdU+ cells in peri-infarct area.
[320] Group A, n = 13: i.c.v. treated with IGF-I; Group B, = 14: placebo; Group D, = 10: s.b. treated with IGF-I; Group E, = 10: placebo; Groups C and F sham-operated controls (= 5 and = 3, respectively). Transient right MCAO for 1 hr I.c.v.: 33.33 mcg IGF-I/d for 3 days, s.c.: 200 mg IGF-I/d for 7 days Treatment was begun 30 min. after MCAO There was less neurological deficit after ischemia in i.c.v. and s.c. IGF-I–treated animals compared with controls. Continuous treatment with i.c.v. and s.c. administered IGF-I achieved a long lasting neuroprotective effect as early as 24 hrs after ischemia.
SDF-1-alfa [255] Rat 16/16 right MCAO ligation and BCCAO Stereotaxical injections with recombinant human SDF-1alfa (4 mcg per one cortical area), vehicle, or control protein (BSA) into 3 cortical areas adjacent to the right MCA At 30 min. after MCA ligation Treated animals showed less cerebral infarction due to up-regulation of antiapoptotic proteins, and they had improved motor performance. SDF-1 alfa injection enhanced the targeting of bone derived cells to the injured brain, as demonstrated in GFP-chimeric mice with cerebral ischemia. Increased vascular density in the ischemic cortex of SDF-1alfa-treated rats enhanced functional local CBF.
[256] Rat and mice From 6 to 12 for different conditions Right MCA ligation and BCCAO Intracerebral injection of human OECs/olfactory nerve fibroblasts (hOECs/ONFs) (1 × 106 cells) into 3 cortical areas adjacent to the right MCA, or vehicle. 1 day after brain ischemia. Cyclosporin A (CsA; 1 g/kg/d, i.p.) injections were given daily to each experimental rat from the day after cerebral ischemia for 3 weeks Transplanted hOECs/ONFs and endogenous homing stem cells colocalized with specific neural and vascular markers, indicating stem cell fusion. Both hOECs/ONFs and endogenous homing stem cells enhanced neuroplasticity in the rat and mouse ischemic brain. The up-regulation of SDF-1α and the enhancement of CXCR4 and PrPC interaction induced by hOEC/ONF implantation mediated neuroplastic signals in response
[246] Male virus- and pathogen-free NMRI mice 27 (comprensive of controls) MCAO After focal cerebral ischemia, SDF-1alfa is up-regulated in the peri-infarct and infarct tissue and this up-regulation was accompanied spatially and temporally by CXCR4-expressing infiltrates.
[247] Mice 40 MCAO Mice received bone marrow transplants from GFP transgenic donors and later underwent a MCAO. SDF-1 expression was detected in the infarcted hemisphere within 24 hr and it is maintained through at least 30 days after MCAO. It was princip ally localized to the ischemic penumbra. At 14 days post occlusion an association between transplanted bone marrow derived cell location and density and the level of SDF-1 immunoreactivity. These findings suggest that SDF-1 has an important role in the homing of bone marrow derived cells, to areas of ischemic injury.
[242] Female Rat 25 2-hr MCAO 3 × 106 male rat BMSCs 1 month after MCAO Treated animals showed significant recovery of behaviour. Expression of SDF-1 was significantly increased along the ischemic boundary zone compared with the corresponding areas in the contralateral hemisphere, suggesting the role of interaction of SDF-1/CXCR4 on the trafficking of transplanted BMSCs.
[257] Rat Naive rats and rats 6 hrs, 1, 2, 4 and 10 days after surgery (n = 3 to 5). Permanent unilateral MCAO or sham operation A strong increase of SDF-1 and CXCR4 expression in infarct and peri-infarct region, 2–10 days after stroke was reported.

MCAO: middle cerebral artery occlusion; i.v.: intravenously; s.c.: subcutaneously; i.p.: intraperitoneally; BBB: blood brain barrier; CCAO: common carotid artery occlusion; SVZ: sub-ventricular zone; i.c.v.: intracerebroventricularly; NSCs: neural stem cells; BMSCs: bone mesenchimal stem cells; BCAO: bilateral carotid artery occlusion; NPCs: neural progenitor cells; CBF: cortical blood flow; BCCAO: bilateral common carotid artery occlusion; SN: substantia nigra; DFO: Deferoxamine; CEPO: non-erythropoietic EPO derivate.

Granulocyte colony-stimulating factor

Human G-CSF is a 19.6 kD glycoprotein encoded by a single gene located on chromosome 17q11–12 [13]. G-CSF contains a hydrophilic signal sequence [14], which is responsible for the mobilization and proliferation of BMSCs, leading to their differentiation in circulating neutrophilic granulocytes. Two variant forms of this protein can be derived from differential splicing of the G-CSF mRNA [15]. G-CSF is produced, above all, by activated monocytes in response to inflammatory cytokines [16], but also by endothelial cells, fibroblasts, mesothelial cells and platelets [17].

Tumour necrosis factor (TNF)-α, interleukin (IL)-1, GM-CSF, IL-4 and bacterial lipopolysaccharide could induce G-CSF production in vitro [1822]. G-CSF functions as a stimulating factor after binding to its receptor (GCSF-R), which is a type I membrane protein consisting of four different domains; an immunoglobulin-like domain, a cytokine receptor-homologous domain and three fibronectin type III domains in the extracellular region [23]. G-CSFR is expressed not only by haematopoietic cells (neutrophils, platelets, lymphocytes and monocytes), but also by endothelial cells, neurons and glial cells [2428]. GCSF-R activates a variety of intracellular pathways, including the Janus kinase (JAK)/signal transducer and activator of transcription (STAT), the Ras/MAPK and phosphatidylinositol 3-kinase (PI3K)/protein kinase B [2932]. Activation of these pathways mediates proliferation, differentiation and survival of hematopoietic cells [33].

The pharmacological effects and side effect profile of G-CSF are well known, since recombinant G-CSF is a licensed drug commonly used in clinical practice to treat chemotherapy-induced neutropenia and to boost stem cell mobilization in bone-marrow transplantation. The most common side effects are bone pain and asymptomatic increases in serum alkaline phosphatase and γ-glutamyl trans-peptidase [34].

G-CSF and experimental stroke

In vitro, G-CSF was reported as being neuroprotective in cell cultures exposed to glutamate-induced excitotoxicity [35] and to promote neuronal differentiation [36] G-CSF was identified as having trophic effects on different cell types within the central nervous system (CNS) and to induce neuroprotection during recovery after brain injury [37]. Endogenous G-CSF seems to play an important role in the brain’s response to ischemia. G-CSF and its receptor are expressed in response to ischemia in the penumbral region of ischemic stroke [36] where it has an anti-apoptotic effect, resulting in reduced cell death in the ischemic penumbra [36, 38, 39]. Data from a post-mortem study demonstrated a strong neuronal G-CSF and GCSF-R immunoreactivity in the infarct and peri-infarct areas in both acute and subacute stroke. Neuronal G-CSF was expressed as a very early response to ischemic stroke, whereas in the subacute phase it was predominantly up-regulated in vascular cells [40]. A clear demonstration of the role of endogenous G-CSF in response to brain ischemia comes from the detection that G-CSF-deficient mice are prone to develop larger infarcts and have worse clinical outcome, and that these effects were reversed after G-CSF administration. In the same study, both real time PCR and immunohistochemistry detected an up-regulation of matrix metalloproteinase 9 (MMP-9), proteins that are normally present in the brain in latent form and that, once activated, contribute to the injury process through neurovascular matrix degradation and increased vascular permeability [41]. This finding also supports a role for GF in the endogenous response to ischemic brain damage [4244].

In an animal model of permanent as well as transient middle cerebral artery occlusion (MCAO), G-CSF produced a significant reduction in brain oedema partly mediated by suppression of injury-induced up-regulation of IL-1, TNF-α and endothelial nitric oxide synthase (eNOS) mRNA [45]. Blood brain barrier (BBB) damage is probably prevented by the decreased production of pro-inflammatory cytokines, the resulting T-cell infiltration [46, 47], and by up-regulation of anti-inflammatory molecules [48] that may be involved in the neuroprotective activity. In addition, G-CSF mobilizes exogenous CD34+ stem cells and promotes their homing to the site of injury in rat brain [49, 50]. After focal cerebral ischemia, G-CSF administration significantly reduced infarct size, mortality and improved neurological function [51, 55].

Two recently published meta-analyses confirm these results. Minnerup et al. reviewed 13 studies evaluating the efficacy of G-CSF in animal models with focal cerebral ischemia. In a total of 277 animals used to determine infarct size and 258 animals used for functional outcome assessments, an overall infarct size reduction of 42% and a functional improvement of sensorimotor deficits ranging from 24% to 40% were observed [56]. G-CSF efficacy in the acute phase (within 6 hrs) after stroke seemed to be dose dependent, and its beneficial effect after delayed administration was confirmed [56]. England et al. [57] included 666 animals from 19 different studies; all these studies, except for those by Lan et al. [58] and Han et al. [35], were included in the previous meta-analysis. Interestingly, the authors further analysed the outcome on the basis of different experimental models demonstrating a significant infarct size reduction after transient but not permanent ischemia [57].

Moreover, G-CSF administration after reperfusion in animal models of focal cerebral ischemia induced by transient (60–90 min.) MCAO was associated with a neuroprotective effect mediated by activation of anti-apoptotic pathways [38, 39, 59] and reduction of focal inflammatory responses [60, 61]. Indeed, phosphorylation of STAT3 and nuclear Pim-1 levels were significantly increased after G-CSF administration, and JAK2/STAT3 pathway activation may represent the anti-apoptotic effect of G-CSF mediated through GCSF-R [36, 38, 39, 51]. Immunohistochemical analysis revealed a significant reduction in cells expressing pro-inflammatory cytokines such as TNF-α, transforming growth factor-β (TGF-β), and inducible nitric oxide synthase (iNOS), and an overexpression of the anti-inflammatory cytokine, IL-10 [61] in peri-ischemic areas [60].

In contrast to these data, a recent study in a murine model of cerebral ischemia showed that G-CSF induces an excessive inflammatory response, resulting in cortical atrophy and impaired behavioural function [62]. There is some evidence that, under ischemic conditions, G-CSF administration results in a significantly increased number of BrdU+ endothelial cells during the acute phase [63] and after 7 days [64, 65]. Also, the combined administration of G-CSF and SCF increased the number of bone marrow-derived endothelial cells in mouse brain after cerebral ischemia [66]. G-CSF may also enhance proliferation and GF production in cultured BMSCs [67]. Taken together, these findings demonstrate that G-CSF promotes both neurogenesis and angiogenesis in peri-ischemic areas. Finally, G-CSF was shown to promote cell proliferation from SGZ of DG, leading to their mobilization and homing to brain [65, 68].

G-CSF and human stroke

The promising results in animal models of stroke led to the rationale for the clinical use of G-CSF, and phase I/II clinical trials were implemented to confirm whether similar responses could be obtained in ischemic human brains. We found three completed trials of G-CSF. The Stem cell Trial of Recovery Enhancement after Stroke was a randomized, controlled, pilot study to assess the safety of G-CSF administration in 36 patients with recent ischemic stroke and to evaluate its effect on circulating CD34+ stem cells. Patients were recruited within 7 to 30 days from symptom onset and then randomized to receive either subcutaneous recombinant human G-CSF or placebo (saline) in a dose-escalation study. The primary outcome was peak circulating blood CD34+ count. Safety assessments included mortality and clinical evaluations with the stroke scales score. This pilot study provided evidence that G-CSF administration in stroke patients is safe, well-tolerated and effective in mobilizing bone marrow CD34+ stem cells, although the benefits on clinical outcome were uncertain [69]. Shyu et al. [70] assessed the safety and efficacy of G-CSF administration in patients with acute ischemic stroke in a small, randomized, blinded, controlled trial in Taiwan. The study was designed to investigate the tolerability and effectiveness of G-CSF administration in acute stroke patients. This trial included 10 patients with acute cerebral infarction (in the MCA territory confirmed by magnetic resonance imaging [MRI]), recruited within 7 days of symptom onset. The primary end-point was clinical improvement at the 1 year follow-up assessed through four different stroke scales; the National Institutes of Health (NIH) Stroke Scale, European Stroke Scale, ESS Motor Subscale (EMS) and Barthel index. As a secondary end-point, the authors used cerebral PET to quantify cerebral uptake of fluorodeoxyglucose (FDG) in the peri-infarct area.

Patients assigned to the treatment arm (n = 7) received 15 μg/kg G-CSF subcutaneously for five consecutive days. All patients underwent a 1–3 month follow-up visit for 1 year and FDG-positron emission tomography (PET) and MRI at 6 and 12 months. Despite the small sample size, this study supports the feasibility of subcutaneous (s.c.) G-CSF administration in stroke patients and suggests that G-CSF may reduce long-term disability. The benefit seemed to be greater in the subgroup of patients who underwent treatment within 24 hrs [70].

Zhang et al. [71] recruited 45 acute stroke patients to a randomized, placebo-controlled trial. Of these, 15 were randomized to recombinant G-CSF (rhG-CSF) 2 g/kg s.c. for 5 days within 30 days of symptoms onset, and 30 were enrolled in the control arm. The primary end-points were NIHSS score and adverse reactions. The authors found a significant reduction of NIHSS score in the treatment group on the 20th day of follow-up (this difference was absent in the placebo group). No adverse reactions were registered.

Recently, the Cochrane Systematic Review, including the three previous small clinical studies, failed to demonstrate a significant reduction in combined death and dependency in stroke patients after G-CSF administration, concluding that further studies are necessary to determine whether G-CSF administration could improve the outcome of stroke patients [72].

A multicenter, randomized, double-blind, placebo-controlled, dose-escalation phase IIA trial (AXIS) was recently completed. In the 45 acute stroke patients in this trial, G-CSF was safe, well-tolerated and showed clinical efficacy, even in patients with wide baseline lesions detected by diffusion-weighted imaging (DWI)-MRI [73].

Ongoing studies

A phase IIB/IIIA trial (AXIS), that will start in the second quarter of 2009 [74], and the Regeneration in Acute Ischemic Stroke Study, were implemented to confirm the safety of G-CSF administration in acute stroke patients [75]. The phase I, granulocyte colony-stimulating factor in ischemic stroke trial is also ongoing to verify the mortality and adverse effects of G-CSF therapy as the primary end-point (http://www.clinicaltrials.gov, NCT00809549). Another phase II clinical trial currently recruiting patients was implemented to assess whether s.c. G-CSF and erythropoietin administration (EPO) influence associative learning and/or motor skills in patients who experience chronic stroke or amyotrophic lateral sclerosis (http://www.clinicaltrials.gov, NCT00298597).

Current evidence and future perspectives

Endogenous G-CSF plays an important role in brain response to ischemia. G-CSF administration in animal models is associated with reduced infarct volume, decreased mortality and better clinical outcome. Recombinant G-CSF is already approved for limited clinical use in human beings. G-CSF administration in stroke patients was safe, but further studies should be implemented in order to demonstrate its efficacy.

Erythropoietin

EPO is a protein hormone of about 34.4 kD and a member of the class I cytokine family [76]. Human EPO is encoded by a single gene consisting of 5 exons located on chromosome 7 [77]. Gene expression is controlled by an oxygen-sensing, hypoxia-inducible factor-dependent mechanism [78]. EPO [79] was shown to increase transcription of cyclins, inhibit cell-cycle inhibitors and to increase the concentration of the anti-apoptotic protein, BCLXL [76, 80], favouring red cell proliferation and promoting the differentiation of bone-marrow stem cells into circulating mature red cells. It is produced by renal interstitial cells in the peritubular capillary beds of adult kidney, and by liver perivenous hepatocytes [8183]. In addition to renal and liver tissues, EPO is also expressed in many extra-renal tissues in cell types such as astrocytes [84], neurons [85] and bone marrow macrophages [86]. It functions as a stimulating factor by binding to a specific erythropoietin receptor (EPOR), a member of the cytokine type 2 family [8789], which is a 66 kD glycoprotein encoded by a gene on chromosome 19. The EPOR protein consists of a single transmembrane domain and an extracellular domain [90, 91].

The EPOR is widely expressed in non-erythroid cells and tissues such as smooth muscle cells [92], myoblasts [93], vascular endothelium [94], heart [95], kidney [96], neuronal cells [97, 98] and brain [99]. EPOR dimerizes upon EPO binding, which increases the receptor’s affinity for JAK2 in the membrane proximal region of the receptor. This results in JAK2/STAT5 protein phosphorylation and activates other signal transduction pathways [91] including the PI3K and extracellular signal-regulated kinase 1 and 2 (ERK1/2) pathways [100102]. Activation of these pathways prevents apoptosis of late erythroid progenitors by promoting their survival, proliferation and differentiation [102]. In human beings, EPO also enhances megakaryocyte maturation by augmenting platelet counts by 10–20% and increasing platelet function [103, 104].

EPO administration is associated with proliferation of erythroid progenitor cells, which, once differentiated into normoblasts, enucleate and leave the bone marrow [17]. Recombinant EPO is widely used in clinical practice for the treatment of anaemia in patients with end-stage renal failure and patients with anaemia induced by chemotherapy or radiotherapy [105]. A large number of EPO structural and functional variants are now available and under development. The development of a neuroprotective variant with absent or reduced haematopoietic functions represents an important challenge [106, 107].

EPO and experimental stroke

EPO and EPOR are constitutively expressed in the central nervous system and play a significant role during brain development [108]. In neurons, EPO and its receptor activate multiple signal transduction pathways including STAT, PI3K/Akt, Ras/ERK1/2, nuclear factor-kB (NF-kB) and intracellular calcium levels [89]. EPO is neuroprotective in cell cultures. This results in a protective and anti-apoptotic effect on hippocampal neurons [109, 110] as well as on cortical neurons [111, 112] in different in vitro studies. Moreover, data from experimental studies provide evidence that EPO and its receptor are overexpressed in response to hypoxia and ischemic damage and have a neuroprotective effect in vitro as well as in animal models and human beings [110113]. EPOR up-regulation occurs first in neurons and endothelial cells in microcirculation after 12 hrs, and is later observed in both neurons and astrocytes. This phenomenon is prominently observed in the penumbra area [85].

The evidence that endogenous EPO inhibition results in more severe brain damage [89] and that EPOR-deficient mice shows increased sensitivity to hypoxia and increased brain apoptosis [91] further support EPO’s major role in the brain’s response to ischemia. Hypoxia-induced EPO expression is mediated by hypoxia inducible factor (HIF), which may be involved in ischemic tolerance [114].

The anti-apoptotic effect of EPO was shown in different animal models. Studies from neonatal rat and mouse stroke models in showed reduced apoptosis through the differential regulation and expression of genes involved in the apoptotic process [115118].

A similar anti-apoptotic effect was observed in a gerbil model of cerebral ischemia [119, 120]. Villa et al. [120] reported a significant reduction in astrocytes, leucocytes and microglia activation as well as decreased production of pro-inflammatory cytokines like TNF-α, IL-6 and monocyte chemoattractant protein 1 in the ischemic area of a MCAO rat model. The same authors reported a similar anti-inflammatory effect consisting of reduced cellular infiltration, apoptosis and white matter damage after treatment with a non-erythropoietic EPO derivate (CEPO) in a rat model of focal cerebral ischemia [121]. EPO administration was also shown to be associated with improved outcome in an experimental model of traumatic brain injury and with reduced brain oedema [122]. A recent paper showed that EPO reduces neurological symptoms in a mouse model of primary brain oedema [123] supporting the evidence that an important component of EPO’s neuroprotective function may be its role in modulating astrocyte water permeability through the regulation of intracellular calcium oscillations, reducing the risk of astrocyte swelling during stroke and other brain insults.

In addition to its anti-apoptotic and anti-oedema effects, EPO exerted pleiotropic effects as an antioxidant [124, 125], anti-inflammatory [120126], glutamate-inhibitory [127], neurotrophic [128] and angiogenic [88] factors, and influenced stem cell differentiation [88, 91, 129131].

A recently published meta-analysis including 16 studies (more than 340 animals) with comparable outcome measures assessing the efficacy of EPO administration after experimental focal ischemia reported a significant neurobehavioural improvement (38% limb function, 37% neuroscore and 37% memory function) and a significant infarct size reduction of 32%. In most of included studies (12), treatment was initiated after ischemia onset. In three studies, EPO was administrated before ischemia and in one study it was evaluated as both a pre- and post-ischemic treatment. A meta-regression of data from six studies supports the evidence that early treatment (within 6 hrs) is most effective. A higher dose also seems to be associated with better outcome. Different EPO derivatives did not differ regarding infarct size reduction and functional improvement [132].

Interestingly, it has been described that the receptors that mediate the neuroprotective effect after ischaemic and traumatic brain injury [133, 134] are different from those expressed by erthroid precursors, [133] and also several EPO variants that do not bind to myeloid cells have a neuroprotective effect [135, 136]. The use of non-haematopoietic EPO analogues could be a promising option in achieving a more specific action and for avoiding systemic side effects such as an increased haematocrit and increased blood pressure values. However, treatment with deferoxamine (DFO), EPO or the combination of DFO and EPO did not protect against grey or white matter damage in the experimental settings used by van der Kooij et al. [118]. Kumral et al. demonstrated a protective effect for EPO on lipopolysaccharide-induced white matter injury in the developing brain [137].

EPO and human stroke

To our knowledge, there are only two published studies evaluating the safety and efficacy of EPO administered to stroke patients. The first trial by Ehrenreich et al. [138] involved 53 patients (13 patients enrolled in a safety study and 40 patients enrolled in a double-blind proof-of-concept trial) with hyperacute ischemic stroke (drug administration within 8 hrs of symptom onset). All patients recruited had an infarct in the MCA area documented by DWI and fluid attenuation inversion recovery MRI in the acute phase. Intravenous infusion of recombinant human (rh)EPO (3.3 × 104 IU/50 ml/30 min.) was started immediately after recruitment and then administered again 24 and 48 hrs later to provide a cumulative dose of 100,000 IU of rhEPO for each stroke patient. MRI data were obtained on days 1, 3 and 18 for each patient. The primary end-points were 30 days NIHSS and Scandinavian Stroke Scale score, and functional outcome on day 30 (Barthel’s index and modified Rankin scale). Secondary end-points were time-dependent fluctuations in neurologic scores and serum markers of injury (S-100B protein) over 30 days, as well as infarct size on days 3 and 18 as assessed by cerebral MRI. EPO was well tolerated and safe in these stroke patients. Moreover, a possible neuroprotective role was suggested by improved clinical outcome (above all, lower Barthel Index score in the treatment arm) and reduced infarct size as well as serum levels of the circulating glial damage marker, S100B in the treatment arm [138].

On the basis of these promising results, a randomized, double-blind, placebo-controlled multicenter study, the German Multicenter EPO Stroke Trial (http://www.clinicaltrials.gov, NCT00604630), with an accrual goal of over 500 patients, was started in 2003 and concluded in 2008, and confirm the results of the first trial [139].

Ongoing studies

As cited above, there is an ongoing phase II clinical trial of G-CSF and EPO in stroke and amyotrophic lateral sclerosis patients (http://www.clinicaltrials.gov, NCT00298597). Two other studies evaluating EPO in combination with human chorionic gonadotropin (hCG) are ongoing. The first is the just begun REGENESIS (US) study, a phase IIB study of hCG and EPO in acute ischemic stroke patients, with the primary objective of assessing the safety and tolerability of hCG and EPO in this patient population (start date, August 2009; www.clinicaltrials.gov, NCT00715364). The second is the active but no longer recruiting REGENESIS (CA), a phase II study to assess neurological outcome in acute ischemic stroke patients treated with hCG and EPO (www.clinicaltrials.gov, NCT00663416).

Current evidence and future perspectives

The neuroprotective effect of EPO was demonstrated in vitro as well as in experimental models of stroke. Data from two small studies in stroke patients suggest that this hormone is safe and that EPO may have a positive effect on both infarct size reduction and clinical outcome. The development of EPO variants that do not have erythropoietic activity represent a further promising and challenging options for stroke treatment.

Granulocyte-macrophage colony-stimulating factor

Human GM-CSF is a 22 kD glycoprotein encoded by a single gene located on chromosome 5q21–5q32 near the IL-3, IL-4, IL-5, IL-9 and MCSF-R genes [140]. It is a 127 amino acid protein, a member of the haematopoietin receptor superfamily, consisting of cytokine-specific α and common β chains [141]. Other than being responsible for the in vitro stimulation of myeloid precursor clonal proliferation [142], GM-CSF was shown to be involved in neutrophils, monocytes, dendritic cell and in macrophage survival and differentiation [143]. GM-CSF is produced by different cell types such as T and B lymphocytes, macrophages, keratinocytes, eosinophils, neutrophils and endothelial cells [144]. GM-CSF function is mediated by activation of the GM-CSF receptor (GMCSFR) which is a cytokine belonging to the type I receptor group. The α-chain of GMCSFR is specific for binding GM-CSF, whereas the β subunit is involved in signal transduction [144]. GM-CSF binding to its receptor regulates cell proliferation through activation of the JAK2/STAT5 and MAPK pathways, but also regulates apoptosis and cell survival through activation of the JAK/STAT and PI3K/Akt pathways [145]. GMCSFR is expressed in CD34+ hematopoietic progenitors, neutrophils, eosinophils, basophils, monocytes, macrophages, microglia, lymphocytes, endothelial cells and mesenchymal cells. It is also expressed in malignancies such us myeloid leukaemia, acute and chronic melanoma, and prostate and lung cancer [146, 147].

The recombinant form of GM-CSF is recognized as a treatment for chemotherapy-induced neutropenia and also employed to enhance the antiviral effects of azidothymidine in HIV patients [148]. Trials have also been conducted to assess GM-CSF use in sepsis, drug-induced neutropenia, HIV infection, acute myeloid leukaemia, aplastic anaemia, myelodysplasia [148] and recently as adjuvant therapy for indolent B-cell lymphoma [149], cutaneous melanoma [150] and chronic lymphocytic leukaemia [151]. The most common dose-related adverse effects are bone pain, erythroderma, weight gain, oedema and inflammation [152].

Increased levels of the pro-inflammatory cytokines IL-8 and GM-CSF were found in the cerebrospinal fluid (CSF) of patients on day 2 after stroke [153], supporting the evidence of a focal immune response in ischemic brain [154].

GM-CSF and experimental stroke

The GMCSFR is broadly expressed in brain regions with a predominantly neuronal pattern in neurons of DG and SVZ [155]. To our knowledge, there are no published studies investigating the neuroprotective effect of endogenous GM-CSF in GM-CSF deficient mouse models. RT-PCR studies reveal that it is expressed in cultured adult stem cells as well as in rodent brain, and that it can enhance neuronal differentiation in adult stem cells in a dose-dependent manner [156].

GMCSFR α gene expression is induced by brain ischemia [155]. Nakagawa et al. [157] investigated the therapeutic effect of intracarotid injection of GM-CSF in a rat model of 1-hr intraluminal MCAO. In the treatment group, a significant reduction in infarct size (from 412 mm3 to 214 mm3, P < 0.005) and a better 48-hr neurological outcome evaluated through Menzie’s neurological scale were observed. A significant infarct size reduction was also reported in a mouse model of common carotid artery occlusion (CCAO) [158]. Recently, Kong et al. [159] confirmed the neuroprotective effect of intraperitoneally administered GM-CSF in a rat model of MCAO. In the treatment group, a significant infarct size reduction and a favourable clinical outcome (rotarod motor test and motor/sensory) were observed. In addition, GM-CSF administration was shown to modulate the expression of apoptosis-related genes (Bcl-2, Bax, caspase 3 and p53) and to reduce programmed cell death.

Data from the study of Nakagawa et al. [157] substantially confirmed the anti-apoptotic function of GM-CSF demonstrated by a reduction in TUNEL+ (apoptotic) cells and by a significant increase in microglia/macrophage (OX-42+ cells) activation in penumbra area. In culture, GM-CSF administration results in JAK2 kinase pathway activation, PI3K/Akt activation, and in a transient activation of Erk 1/2 that finally results in a substantial anti-apoptotic effect in cortical neurons [159]. In primary cortical neurons and human neuroblastoma cells, GM-CSF counteracted apoptosis and induced Bcl-2 and BCLXL in a dose- and time-dependent manner. Inhibition of the PI3K-Akt pathway resulted in a severe reduction in anti-apoptotic activity [155]. The substantial neuroprotective and anti-apoptotic properties of GM-CSF were demonstrated in vitro as well as in in vivo experimental stroke models in which administration of GM-CSF led to a decrease in the extent of neuronal apoptosis by modulating the expression of several apoptosis-related genes such as Bcl-2, Bax, caspase 3 and p53 [159]. After neonatal hypoxia in rats, GM-CSF treatment decreased apoptotic cells (TUNEL+ cells), reduced expression of Bax, cleaved caspase-3 and increased the expression of STAT3, Bcl-2 and Pim-1, supporting a neuroprotective effect of GM-CSF administration [160].

Buschmann et al. [161] first demonstrated an improvement in brain hemodynamic parameters after GM-CSF treatment in a rat model of cerebral ischemia due to the occlusion of the vertebral plus the left carotid artery [3-VO]. GM-CSF induced a posterior cerebral artery diameter enlargement and enhanced mononuclear cell invasion (macrophages) at the site of vascular collateral proliferation. In other studies in the same experimental model, GM-CSF was shown to protect against hypotension-induced cerebral blood flow (CBF) decline and hypotension-induced ATP depletion, resulting in a reduction in metabolic injury [162].

In a rat model of sequential bilateral carotid artery occlusion, GM-CSF treatment induced restoration of impaired cerebral hemodynamic and favoured structural changes in the resistance-vessel network. In the treatment arm, there was a significantly higher number of intraparenchymal vessels per section than in control groups as well as a higher density of leptomeningeal vessels per section. Moreover, one week after artery occlusion, CD-68+ cell numbers were higher in the perivascular area of GM-CSF treated animals [163]. The more recent report from Todo et al. [158] confirmed the role of GM-CSF in enhancing leptomeningeal collateral growth in a mouse model of unilateral CCA occlusion. In the same study, a significant infarct size reduction in the GM-CSF group was detected.

GM-CSF and human stroke

Despite experimental evidence for the role of GM-CSF in inducing vascular remodelling, restoration of cerebral hemodynamic and mobilization of CD34+ cells [164, 165] after cerebral ischemia, no clinical trials have been implemented or are ongoing to date. A small, recently published study in patients with acute stroke showed that stroke is associated with higher plasma levels of GM-CSF but failed to find an association between GM-CSF levels and better neurological outcome. This finding supports a role for GM-CSF in response to brain ischemia, but further studies are necessary to better clarify its function [166].

Current evidence and future perspectives

In vitro, GM-CSF enhances neuronal differentiation and prevents apoptosis. A substantial anti-apoptotic effect was confirmed in animal models in which GM-CSF administration resulted in improved functional outcome and brain hemodynamic, and infarct size reduction. Despite these promising results and that GM-CSF is already licensed for various clinical uses, studies in stroke patients have not yet been implemented. For its role in remodelling the vascular architecture, GM-CSF may be a suitable candidate for future stroke therapy.

Vascular endothelial growth factor

VEGF or VEGF-A is a 23 kD glycoprotein belonging to a family of heparin-binding GFs including VEGF-B, VEGF-C, VEGF-D and placental-like GF. VEGF is encoded by a gene on chromosome 6 and can be alternatively spliced into at least five different isoforms in human beings (VEGF121, VEGF145, VEGF165, VEGF189 and VEGF206). The mouse VEGF-A isoform has one less amino acid [167] and is responsible for endothelial cell proliferation, migration and survival [168]. First discovered for its regulatory function in vascular endothelial cell permeability, it is now a well-known angiogenic factor with a recognized role in vasculogenesis and vascular maintenance in all mammalian organs.

VEGF is a multifunctional protein involved in capillary regulation in adult skeletal muscle, angiogenesis, inflammation, cancer and wound healing and repair. It is also expressed in epithelial cells in the kidney and lung [167]. VEGF is ubiquitously expressed in brain, mostly by choroid plexus epithelial cells but also by astrocytes and neurons [169, 170].

Many different factors, including both genes and gene products, are known to enhance VEGF production. VEGF is secreted by tumours in response to hypoxia to stimulate angiogenesis to increase oxygen supply [171], but its production is also triggered by oncogenes (c-Src, Bcr-Abl and Ras oncogenes), tumour suppressor genes (e.g. p53) [172175] and other GFs and cytokines such as cyclooxygenase-2 [176] and platelet derived growth factor (PDGF) [177].

VEGF’s biological functions are mediated by its binding to receptors on vascular and lymph-vessel endothelial cells as well as on other cell types including fibroblasts, smooth muscle cells, haematopoietic stem cells, epithelial cells, monocytes and macrophages [178]. Three VEGF receptors, VEGFR-1 (flt-1), VEGFR-2 (KDR/flk-1) and VEGFR-3 (flt-4) with tyrosine kinase activity have been identified. Co-receptors, including heparin sulphate proteoglycans and neuropilins, may modify the tyrosine kinase activity of VEGFR or bind VEGF directly.

VEGFR-1, which binds VEGF-A, B and PLGF, regulates vascular development and signal transduction mediated through ligand-specific patterns of receptor phosphorylation [179]. VEGFR-2, which binds VEGF-A, is important for angiogenesis; VEGFR-3, which binds VEGF-C and VEGF-D, influences lymphangiogenesis.

Neuropilin co-receptors (Np-1 and Np-2) influence vascular endothelial and non-endothelial cells (i.e. podocytes), the protection of neuronal cells, axon growth and guidance, and modulate VEGF distribution in various tissues [180].

VEGF is actually considered one of the most important angiogenesis regulators and a new key target in anti-cancer treatment. The role of anti-VEGF or anti-VEGFR therapy is supported by clinical trials. Currently, the humanized monoclonal antibody, bevacizumab, which binds to VEGF-A, and the small molecule kinase inhibitors sunitinib and sorafenib (which inhibit the kinase activities of VEGFR) were approved for use in patients with various malignancies.

VEGF and experimental stroke

In vitro, VEGF protects cultured motor neurons from hypoxia, reactive oxygen species, and glutamate-induced toxicity [181, 182] and acts as a trophic factor for neuronal stem cells [183]. Moreover, VEGF and VEGFR expression were induced in brain after global ischemia [184] or permanent [185] or transient MCAO, with a different temporal profile among these cells [186188].

In hypoxic and ischemic brain, VEGF expression is induced by transcriptional activation via HIF-1 and HIF-2 [170, 185, 189, 190]. Within 6 hrs after ischemic injury, VEGF expression was increased in the penumbra area [185]. VEGF, VEGFR2 and HIF-1, through their angiogenic functions, seem to be protective in long-term responses to hypoxic injury, even if, in the acute phase, their expression favours oedema formation [191].

VEGF-A was also expressed during hypoxic preconditioning in adult mouse [192]. Immunohistochemistry of brain tissue from adult rats subjected to photothrombotic ring stroke with spontaneous reperfusion showed that the VEGF protein subtypes A and C increased simultaneously 2 hrs after irradiation of the lesion, and from 24 to 72 hrs after stroke in the penumbra-like zone. The expression of VEGF A, C, and their respective receptors (flt-1, flk-1 and flt-4) was also associated with microvessel formation [193]. Since VEGF plays a positive role in angiogenesis, remodelling and re-vascularization of damaged tissues [191] GF has recently been under investigation in ischemic brain [194].

eNOS expression after brain ischemia following permanent MCAO was found to temporarily and anatomically co-localize with VEGF expression mostly in the penumbra area where these proteins probably exert a protective effect on damaged brain tissue [195]. Furthermore, in a rat model of focal embolic cerebral ischemia, exogenous nitric oxide administration was shown to enhance angiogenesis and vasculogenesis by activating the nitric oxide/cyclic guanosine monophosphate pathway and by increasing VEGF levels in ischemic boundary regions [196]. Mice overexpressing the human VEGF165 isoform exhibited a physiologically higher density of brain vessels and enhanced angiogenesis after brain ischemia. Mice overexpressing VEGF also had a smaller infarct area, which correlated with better clinical outcome [197, 198].

VEGF overexpression was associated with a higher capillary density, but not with an increase in blood flow, probably because circulating blood is stolen by the non-ischemic areas [197]. Another experimental model in which recombinant human VEGF165 was infused into the right lateral ventricle of rats 1 week before MCAO showed that VEGF was able to decrease infarct volume and brain oedema without increasing CBF [199].

Several experimental studies substantially confirmed the positive effect of VEGF in ischemic stroke. Transient MCAO is the most common model despite a great deal of variability between different protocols in terms of the administration route (see Table 1).

VEGF administration, as previously reported for other GFs, was associated with an infarct size reduction ranging from 34% to 47%[199202] and improved clinical outcome [198, 202205] without an increase in brain oedema [199, 202]. The relationship between VEGF and brain oedema has been investigated by studies aimed at clarifying the possible underlying mechanisms of brain oedema. The evaluation of VEGF and angiopoietin (Ang-1 and -2) expression in a rat model of irreversible MCAO by Beck et al. [206], showed that, in the early phase after ischemic damage, Ang-2, which plays a role in vascular remodelling by blocking the constitutive expression of Ang-1, is hypoxia induced. Thereafter, Ang-2 expression is likely to follow VEGF up-regulation, since VEGF protein was first detected after 6 hrs with expression peaking after 24 hrs [206]. The combined effects of VEGF and Ang-2 on cerebral angiogenesis was confirmed by Zhu and colleagues, who also reported that Ang-2 and VEGF act in combination in BBB disruption [207]. This effect on the BBB was previously reported in a rat model of focal cerebral ischemia in which MRI) showed that post-ischemic (1 hr) administration of rhVEGF165 significantly increases BBB leakage, haemorrhagic transformation and ischemic lesions, whereas VEGF administration after the acute phase (48 hrs) was associated with angiogenesis enhancement and functional improvement [203].

The link between VEGF and BBB after stroke was further investigated by Zhang et al. in a rat model of embolic stroke where VEGF mRNA was found to be increased 2 to 4 hrs after stroke, whereas Ang-1 mRNA decreased [208]. There was an association between VEGF up-regulation and BBB leakage in the ischemic core. VEGF/VEGFRs and Ang/Tie2 were detected at the boundary of the ischemic lesion from two to 28 days after stroke in association with newly formed vessels. These data support a possible role of Ang-1 in limiting VEGF-induced BBB leakage during the acute phase, whereas up-regulation of VEGF/VEGFR and Ang/Tie2 at the boundary zone may regulate neovascularization and angiogenesis in ischemic brain tissue [206208]. Zhang and Chopp administered exogenous VEGF in a mouse model of focal cerebral ischemia showing that increased levels of Ang-1 reduces BBB leakage [209]. Mesenchymal stem cell (MSC) infusion 24 hrs after MCAO significantly decreased BBB leakage and promoted angiogenesis by increasing endogenous Ang1 and Tie2, and occluding cerebral expression in the ischemic border. Capillary formation was reduced after inhibition of Flk1 or Ang-1 and down-regulation of Tie-2, suggesting a critical role for VEGF/Flk1 and Ang1/Tie2 in MSC-induced angiogenesis [210].

In a mouse model of brain ischemia, inhibition of VEGF activity using a neutralizing antibody was shown to completely block hypoxia-induced vascular permeability [211]. A similar effect was obtained by employing the VEGF antagonist in a rat model of vasogenic oedema due to cerebral venous ischemia [212]. In a 90-min. MCAO rat model, pre-treatment with a KDR kinase inhibitor elicited an early, transient decrease in oedema and subsequent reduction in infarct volume, implicating VEGF as a mediator of stroke-related vascular permeability and ischemic injury [201].

The role of VEGF in stroke-oedema is confirmed by further experimental models. Stereotactic injection of VEGF into the left substantia nigra (SN) resulted in highly up-regulated aquaporin-4 (AQP4), which is the most abundant water channel in the brain. AQP4 mRNA and protein in the ventral midbrain localized in close proximity with the new VEGF-induced blood vessels suggest a positive role of AQP4 in oedema resolution, which may partially explain the previously reported beneficial effects of delayed VEGF administration in ischemic rats [213].

In a gerbil model of focal brain ischemia, pre-treatment (6 days or 12 days) with an intracerebroventricular injection of adeno-associated virus transferring the gene for VEGF (rAAV-VEGF) improved survival and brain oedema, and delayed neuronal death [214]. However, this method of administration is controversial with some authors reporting that intraventricular rAAV-VEGF pre-treatment in the transient MCAO rat model can result in deleterious intracranial hypertension and augment secondary ischemic insults during the early stages. They concluded that pre-ischemic VEGF gene transfer via an intraventricular approach may not be a favourable therapeutic strategy for MCAO, which should be adopted with caution or avoided in experimental stroke [215].

Also, different dosages of VEGF165 can injure rather than promote recovery of nervous tissue [216]. In this experimental model, VEGF165 was administered intra-arterially at different dosages in temporary MCAO rats. In this study, brain treated with low non-angiogenic doses of VEGF165 showed preserved neuropils and reduced numbers of macrophages in comparison with ischemic brains treated with high angiogenic doses, which showed phagocytized neuropils and a high macrophage density.

Both HIF-1α and VEGFR-2 were implicated in peri-infarct neuroprotection. In squirrel monkeys, a bipolar electro-coagulation of cortical vessels supplying the M1 hand area representation was performed. It was shown that HIF-1a up-regulation was confined to the infarct and peri-infarct regions. Increases in VEGFR-2 immunoreactivity occurred in two remote regions. Thus, while remote areas undergo a molecular response to the infarct, the authors suggested that there is a delay in the initiation of the response, which may ultimately increase the ‘window of opportunity’ for neuroprotective interventions in the intact cortex [217].

Regarding new insights in VEGF mediated angiogenesis, MCAO was performed in mice overexpressing (VEGF-hi/+) and underexpressing (VEGF-lo/+) VEGF. Compared with VEGF-hi/+ mice, VEGF-lo/+ mice formed fewer collaterals during the perinatal period when adult density was established, and had a 2-fold larger infarction after MCA ligation, suggesting that VEGF and VEGFR-1 are determinants of arteriogenesis. VEGF-A specifies formation of native collaterals in healthy tissues [218].

In cerebral ischemia, Chu et al. investigated the effects of combined intravenous administration of transplanted neural stem cells and VEGF on focal cerebral ischemia in rats. Combined cell and pharmacological therapy was associated with better functional recovery (improved sensorimotor deficit and better performance on the behaviour tests), reduced cerebral atrophy and increased vascular density without a significant effect on stem cell survival [205].

After transplantation of neural progenitor cells (NPCs) expressing VEGF-A165 into the caudate putamen in rats, Maurer et al. [219] found an increased survival of adenovirally transfected NPCs and an increased expression of the endothelial cell marker, PECAM-1 (CD31) demonstrating that the graft itself is a useful vehicle for GF delivery, promoting the survival of NPCs. Moreover, transplantation of VEGF-expressing NPCs supports angiogenesis in the brain, which may contribute to brain repair.

Intracerebral administration of VEGF gene-transferred BMSCs engineered with a replication-deficient herpes simplex virus type 1 1764/4-/pR19-hVEGF165 vector in a rat model of MCAO determined a more significant functional recovery, decreased infarct volume and showed stronger expression of VEGF proteins compared with transplantation of native BMSCs alone [220]. Moreover, the neuroprotective functions of VEGF were also mediated by its anti-apoptotic role as shown by a significant reduction in TUNEL staining in the VEGF-treated group, both at 24 and 48 hrs in the study of Hayashi et al. [200].

VEGF and human stroke

No clinical studies with VEGF have been implemented so far in ischemic stroke patients.

Ongoing studies

One study is active but not ongoing. It focuses on angiogenesis after intracardiac injection of VEGF (phase I) and oral L-arginine supplementation (phase II) in coronary artery disease patients. This study could provide useful information on the effects of VEGF on angiogenesis after ischemic injury (www.clinicaltrial.gov, NCT00134433).

Current evidence and future perspectives

VEGF is a trophic factor for neuronal stem cells and its expression increases after cerebral ischemia. In animal models, VEGF administration results in better functional outcome and reduced infarct size and oedema. Currently there are no data from studies assessing the safety and efficacy of VEGF in stroke patients.

Stromal cell-derived factor-1

SDF-1 is a CXC chemokine produced by bone marrow stromal cells. Two isoforms, SDF-1α and SDF-1β, arising from alternative splicing of the SDF-1 mRNA, are identified. The resulting proteins, SDF-1α and SDF-1β are identical except for four C-terminal amino acids and represent endogenous ligands for the CXC chemokine receptor 4 (CXCR4), which also functions as co-receptor for HIV-1 in lymphocytes [221223]. SDF-1 is a potent chemoattractant for hematopoietic stem cells (HSCs) and CD34+ cells, which express CXCR4. Thus, SDF-1α, which is constitutively expressed in all tissues, plays an important role in HSCs trafficking between the peripheral circulation and bone marrow [224, 225]. In brain, SDF-1/CXCR4 is recognized as being expressed in a variety of neuronal tissues during development, including cerebellum [226], cerebral cortex [227, 228], DG [229] and motor axons [230], where it was shown to regulate synaptic transmission and neural cell migration [231, 232] and to guide migration of neural progenitors from the external germinal layer to the internal granular layer [233235]. The detection of increased SDF-1α expression in the brain of HIV-positive patients supports the notion that SDF-1α may also play a role in neuroprotection and neuronal plasticity [236]. SDF-1 up-regulates the expression of VEGF in neurovascular endothelial cells [237]. In addition, the SDF-1α/CXCR4 axis is related to arginine vasopressin release in adult brain [238240] and microglia-enhanced, SDF-1-stimulated, TNF-β and glutamate release from astrocytes in vitro. This mechanism is also proposed as a novel excitotoxic pathological mechanism in some neurodegenerative diseases [241].

Expression of both SDF-1α and CXCR4 was up-regulated, mostly under hypoxic conditions such as acute renal failure, ischemic cardiomyopathy and ischemic brain injury [242244].

Liu et al. reported that synchronized Ca2+ spikes among cultured hippocampal neurons, which represent periodic burst firing of action potentials believed to play a major role in the development and plasticity of the neuronal circuitry, can be modulated by two small factors that act on G-protein-coupled receptors (GPCRs): the neuropeptide pituitary adenylate cyclase-activating polypeptide, and the chemokine SDF-1. This modulation of neuronal activity through GPCRs may represent a significant mechanism that underlies the neuronal plasticity during neural development and functioning [245].

Stumm et al. (2002) demonstrated that an isoform-specific regulation of SDF-1, obtained by alternative splicing, modulated neurotransmission and cerebral infiltration through distinct CXCR4 pathways. After focal ischemia, SDF-1β expression increased in the endothelial cells of the penumbra blood vessels and decreased in non-lesioned brain areas, whereas there was concomitant infiltration of CXCR4-expressing peripheral blood cells. In the same experimental model, neuronal SDF-1α was transiently down-regulated and neuronal CXCR4 was transiently up-regulated in the non-lesioned cortex after ischemia. Thus, whereas SDF-1β may control cerebral blood cell infiltration, the neuronal SDF1-α/CXCR4 system may contribute to ischemia-induced neuronal plasticity, suggesting that the isoform-specific regulation of SDF-1 expression modulates neurotransmission and cerebral infiltration [246].

SDF-1 in experimental stroke

Stumm et al. [246] showed that SDF-1 is up-regulated in peri-infarct and infarct tissue in a mouse stroke model, and that this up-regulation was accompanied spatially and temporally by CXCR4-expressing infiltrates.

The results of this study were confirmed in other experimental models. Hill et al., using anti-SDF-1 antibodies, detected increased expression of SDF-1 protein in MCAO stroke rats [247]. In this study, SDF-1 expression was observed in infarcted hemispheres within 24 hrs, increased by 7 days after MCAO, and was maintained for at least 30 days after occlusion of the MCA. SDF-1 expression was principally localized to ischemic penumbra, and appeared to increase as one moved from the periphery of the penumbra in towards the border of the infarct core. The prominent expression of SDF-1 in the penumbra area when new blood vessels are forming supports the role of SDF-1 in brain remodelling after ischemic injury. The authors also reported an association between transplanted bone marrow derived cell location and density and the level of SDF-1 immunoreactivity on day 14 after occlusion. These findings suggest that SDF-1 has an important role in bone marrow-derived cell homing, especially monocytes, to areas of ischemic injury.

The extended expression of SDF-1 in the penumbra, and at a later time in the ischemic core when new blood vessels are forming, is concordant with its being involved in some aspects of brain remodelling following ischemic injury.

Thus, there is considerable evidence for SDF-1s having a role in targeting neuronal precursors as well as neural and bone marrow-derived MSC towards an ischemic brain lesion [242, 248253]. CXCR4 was identified in tissue-committed bone marrow stem cells, which express neural lineage markers and may become attracted to ischemic brain tissue via SDF-1 [254].

The role of this chemokine as a signal adhesion molecule and in inducing migration of HSCs to injured brain tissue in an ischemic animal model was further demonstrated by Shyu et al. [255]. These authors injected recombinant SDF1α protein intracerebrally into MCAO rats and treated cells in culture with SDF-1. This treatment increased engraftment of BrdU-labelled bone marrow derived cells in the ipsilateral cortex near the ischemic area boundary and in the sub-ventricular region of the ischemic hemisphere.

The same authors performed an intracerebral injection of human olfactory ensheathing cells (OECs)/olfactory nerve fibroblasts (hOECs/ONFs) in a stroke model. Transplanted and endogenous homing stem cells co localized with specific neural and vascular markers, suggesting a stem-cell fusion phenomena. Interestingly, both hOECs/ONFs and endogenous homing stem cells enhanced neuroplasticity in rat and mouse ischemic brain models by up-regulation of SDF-1α and CXCR4 [256].

A great increase in SDF-1 and CXCR4 expression in the infarct and peri-infarct regions 2–10 days after stroke was also recently reported by Schonemeier et al. [257] in MCAO rats. These results support the possible role of a SDF-1 a/CXCR4 interaction in adaptive reorganization and inflammation of post-ischemic injury other than in vascular remodelling, angiogenesis and neurogenesis, thereby alleviating stroke symptoms. Since the infarct volume was significantly reduced in SDF-1α treated rats compared with controls 3 days after stroke, the authors suggested that SDF-1α may provide a neuroprotective function against neurotoxic insult and exert an anti-apoptotic effect [257]. To our knowledge, SDF-1 has never been tested in stroke patients.

Current evidence and future perspectives

SDF-1 is up-regulated in different hypoxic conditions and acts mostly as a signal adhesion molecule in inducing and targeting cell migration and homing towards ischemic lesions. To our knowledge, SDF-1 has never been tested in stroke patients. However, improvement in locomotor activity tests and the reduction of infarct volume 3 days after stroke observed in rats treated with SDF-1 could support the use of SDF-1 in inducing brain plasticity and functional recovery.

Stem cell factor

SCF is an essential haematopoietic cytokine that interacts with other cytokines to preserve the viability of hematopoietic stem and progenitor cells, influencing their entry into the cell cycle and facilitating their proliferation and differentiation. When combined with other cytokines, SCF increases the cloning efficacy of hematopoietic progenitor cells from all lineages and the clonogenic activity of other CSFs [258].

SCF alone cannot drive non-cycling haematopoietic progenitor cells into the cell cycle, but can prevent these cells from undergoing apoptosis. Particularly, SCF and G-CSF favour neurogenesis and have a neuroprotective effect partly mediated by up-regulation of anti-inflammatory cytokines such as IL-10 [61]. Moreover, SCF plus G-CSF augment bone marrow derived cells that express the neuronal marker, NeuN in intact brains [259] as well as in acute and subacute ischemic brains [59]. The combined use of these GFs also supports the mobilization of BMSCs into the brain and their differentiation into neuronal cells. SCF and G-CSF administration, in the acute phase after focal ischemia induced by permanent MCAO, was associated with infarct volume reduction [59]. In a rat model, SCF administered in the acute phase and within the 7 days after experimental stroke was associated with enhanced neuronal progenitor cell proliferation, reduced infarct size and improved clinical outcome. In this study, functional improvement was delayed in SCF-treated animals, but was long lasting compared with G-CSF-treated animals [260]. The same authors demonstrated a beneficial effect of SCF treatment in an animal model of chronic brain ischemia [260, 261] showing that systemic administration of SCF plus G-CSF during the chronic phase of brain ischemia, initiated 3.5 months after ischemia for 7 days, led to improved functional recovery with a reduced size of the infarct cavities. However, the mechanisms by which SCF plus G-CSF help to repair the damaged brain during chronic stroke remains unknown. Administration in the subacute phase similarly reduced infarct area, but also promoted the generation of neuronal cells from both bone marrow derived cells and intrinsic neural stem/progenitor cells, resulting in improved functional recovery [261].

Current evidence and future perspectives

This observation indicates that combined SCF and G-CSF treatment may favour mobilization of BMSCs into the brain and their differentiation into neuronal cells in both acute and chronic stroke, leading to improvement of functional recovery with reduced infarct size. No studies on the use of SCF alone are available. Despite the absence of available clinical trials, the results of experimental studies provide interesting insights into the use of SCF and G-CSF in chronic stroke.

Other growth factors: neurotrophins

It has been observed that cerebral ischemia up-regulates expression of several GFs such as brain–derived neurotrophic factor (BDNF), fibroblast growth factor (FGF)-2 and IGF-1, epidermal growth factor (EGF) and glial cell line-derived neurotrophic factor (GDNF) [262].

Neurotrophins are a family of structurally related polypeptides that play a critical role during neuronal development and appear to mediate a protective response in mature animals [263]. Neurotrophins demonstrated a protective role in several experimental stroke studies (Table 2). Human mesenchymal stem cells (hMSCs) have the capacity to secrete neurotrophins in culture [264267]. hMSCs respond to the ischemia-injured brain environment through an increased production of neurotrophins and angiogenic factors [268].

Table 2.

Neurotrophins experimental studies

Neurotrophins Study reference Animal model Sample size (treatment group/control group) Type of study (focal or global ischemia) Treatment Time of administration Results
IGF-1 [318] Rat N° Experiments: n° animals; 1: 20 mcg IGF-1 (= 18); 1: 20 mcg IGF-2 (= 20); 1: 20 mcg des-IGF-1 (= 18); 2: Vehicle (= 12); 2: 2 mcg des-IGF-1 (= 12); 2: Vehicle (= 26); 2: 150 mcg des-IGF-1 (= 26); 3: Vehicle (= 20); 3: 20 mcg IGF-1 (= 20); 3: 20 mcg IGF-1 + 30 mcg IGF-2 (= 20); 4: [3H]IGF-1/20 mcg IGF-1 (= 4); 4: [3H]IGF-1/20 mcg IGF-1 + 30 mcg IGF-2 (= 4) Right CCA ligation, and after recovery from anestesia and 10 min. of inhalational hypoxia I.c.v. infusion of recombinant human IGF-1 and/or IGF-2, at different dosages, or vehicle alone. 2 hrs after hypoxia 20 mcg IGF-1 reduced neuronal loss in all regions. Neither 20 mcg IGF-2, 2 mcg des-IGF-1, nor 20 mcg des-IGF-1 reduced neuronal loss. There was a trend towards a reduction in neuronal loss after 150 mcg des-IGF-1. IGF-2 alone increased neuronal loss in the hippocampus and DG compared with vehicle-treated animals. Coadministration of 30 mcg IGF-2 blocked the neuroprotective effects of 20 mcg IGF-1 and reduced the accumulation of F3HJIGF-1 in the injured hemisphere.
[320] Group A, n = 13: i.c.v. treated with IGF-I; Group B, = 14: placebo; Group D, = 10: s.b. treated with IGF-I; Group E, = 10: placebo; Groups C and F sham-operated controls (= 5 and = 3, respectively). Transient right MCAO for 1 hr I.c.v.: 33.33 mcg IGF-I/d for 3 days, 200 mg IGF-I/d for 7 days Treatment was begun 30 min. after MCAO There was less neurological deficit after ischemia in i.c.v. and s.c. IGF-I–treated animals compared with controls. Continuous treatment with i.c.v. and s.c. administered IGF-I achieved a long lasting neuroprotective effect as early as 24 hr after ischemia.
[322] Mice 8 for each group 3 weeks after gene transfer the mice underwent permanent distal MCAO. Long-term cerebral IGF-1 overexpression by the AAV transduction system through stereotaxic injection. Control mice were injected with AAV–green fluorescent protein or saline. IGF-1 gene transfer compared with control treatment significantly improved motor performance, demonstrated reduced volume of cerebral infarction. IGF-1 gene transfer potently increased neovessel formation in the peri-inf arct and injection needle tract area compared with AAV–GFP transduction. Increased vascular density was associated with increased local vascular perfusion. AAV-IGF-1 treatment enhanced neurogenesis in the SVZ compared with AAV–GFP treatment.
[323] Diabetic animals MCAO i.v. infusion of IGF-1 Acute administration of IGF-1 30-min. before or 2 hr after MCAO followed by 24-hr reperfusion Significant decrease of both lesion volume and apoptosis
[317] Rat n = 3/time-point/set: -1, 5 and 10 hrs, 1, 3, 5 and 10 days after 60 min. hypoxia -1, 3 and 5 days after the 15-min. hypoxia Right CA ligation. Hypoxia for 15 or 60 min. Authors found cell type-specific expression for IGF-1, IGFBP-2, 3, 5 and 6 after injury. IGF-1 produced by microglia after injury is transferred to perineuronal reactive astrocytes expressing IGFBP-2
[302] Mice and LID mice 30 min. MCAO human IGF-I (hIGF-I, 50 μg/kg/day, by a s.c. long-term delivery system Before MCAO Authors found that chronic high serum IGF-I correlates with increased brain infarct size following MCAO, while low levels correlate with reduced lesion size, suggesting that lowering serum IGF-I levels in aging mammals, may beneficial against the increased risk of stroke associated to old age.
[319] Late gestation fetal sheep 6 per group 30 min. bilateral inflation of carotid artery 0.1 mcg rhIGF-1, 1 mcg hIGF-1, 10 mcg rhIGF-1, or vehicle was infused into a lateral cerebral ventricle over 1 hr. 2 hrs after MCAO Overall neuronal loss was reduced with 0.1 mcg and 1 mcg rhIGF-1, but treatment with 10 mcg was not effective. With 1 mcg rhIGF-1 neuronal loss scores were significantly lower in cortex, hippocampus, and striatum, 1 mcg rhIGF-1 also delayed the onset of seizures and reduced their incidence.
[321] Rat 150 μg rhIGF-I (n = 10); 37.5 μg rhIGF-I (n = 10); controls (n = 9). Permanent MCAO Intranasal delivery of vehicle-control, 37.5 and 150 μg of recombinant human IGF-I (rhIGF-I) At 10 min. after onset of 2 hrs of MCAO, and then 24 and 48 hr later Treatment with the 150 μg IGF-I significantly reduced the infarct volume versus control and improved all the neurologic deficits. The 37.5-μg dose of IGF-I was ineffective.
GDNF [333] rLV-GFP, n = 20+5; rLV-GDNF, n = 12+4; non-transduced, n = 6; rAAV-GFP, n = 8; rAAV-GDNF, n = 8 30 min. of MCAO 1° experiment: intrastriatal injections of rLV carrying the gene for GFP (rLV-GFP, or GDNF (rLV-GDNF) or were given an incision of the skin overlying the skull only; 2° experiment: rats were given injections of rAAV carrying the gene for GFP (rAAV-GFP, or GDNF (rAAV-GDNF), into the SN 1° experiment: 4 weeks prior to MCAO or sham surgery; 2° experiment: 6 weeks prior to 30 min. of MCAO The two routes of GDNF gene delivery both effectively promoted high striatal GDNF levels, which persisted several weeks after the stroke. Contrary to previous experiments, the treatment did not increase neuron survival or improve functional recovery. The authors proposed that the net effect of is complex and may depend on several experimental factors
[329] Rat 9+7/7+5+6 controls MCAO GDNF (2.5 mcg) or vehicle were placed in contact with the surface of the cerebral cortex. After MCAO Both infarct size and brain edema after permanent MCAO were significantly reduced by topical application of GDNF. Induction of TUNEL staining and immunoreactivities for caspases-1 and -3 was greatly reduced with GDNF treatment.
[330] Rat 59 40 min. transient middle MCAO and BCCAO Human recombinant GDNF was injected initially i.c.v. (0.4 mcg/mcl × 10 mcl). Three injections of GDNF (0,4 mcg/μl × 5 mcl X 3 sites) were made directly into the cortex adjacent to the MCA. 5 min. after intracerebral GDNF Injection, the left MCA and bilateral CCAs were ligated for 40 or 90 min. in the aged and young animals, respectively. GDNF protects the cerebral hemispheres from damage induced by MCAO. The increase in nitric oxide that accompanies MCAO and subsequent reperfusion is blocked almost completely by GDNF.
[331] Rat Vehicle = 5+4+2; Ad–LacZ = 5+4; Ad–GDNF 6+4+2; 1 hr after Ad–GDNF = 6+4+2; Sham control sections = 2 90 min. of transient MCAO Ad–LacZ or vehicle solution was administered to the ipsilateral cortex . Ad–GDNF (10 μl). Just, or 1 hr after reperfusion Ad–GDNF significantly reduced the infarct volume when immediately administered after the reperfusion, but became insignificant when administered at 1 hr after the reperfusion. The protective effect of GDNF was related to the significant reduction of the number positive cells positive cells for active caspase-3 but not -9.
[332] Rat Rats were killed after 2, 6, 24 hrs, or 1 week of reperfusion (n = 5 for group). Sham surgery only = 6 Either 30 min. or 2 hrs of MCAO Authors found major changes of GDNF family signaling in the forebrain, regulated mainly through altered receptor levels, in the post-ischemic phase that could enhance neuroprotective and neuroregenerative responses both to endogenous and exogenous GDNF ligands.
[334] Adult spontaneously hypertensive rats. 1 hr transient MCAO i.c.v. IGF-1 and GDNF infusions by osmotic minipump. BrdU (i.p.; twice a day; 50 mg/kg starting at 1 day after MCAO or sham surgery 1 day before of MCAO MCAO-induced progenitor cell proliferation in the ipsilateral DG was significantly increased by i.c.v. infusion of IGF-1 and GDNF compared to vehicle.
[335] 1° GDNF (n = 9) vehicle (n = 9) 2°: GDNF (n = 10) vehicle (n = 10) 2-hr MCAO 1° experiment: Recombinant human GDNF (1.5 mcg/μL) or vehicle was infused intrastriatally via osmotic minipumps. BrdU, 50 mg/kg was injected i.p. 3 times at 2-hr intervals on day 7 2° experiment: BrdU was given 3 times daily for 1 week starting 6 days after the 2-hr MCAO. 1° experiment: after MCAO for 7 days. 2° experiment: from day 13 to day 26 after MCAO. GDNF infusion increased cell proliferation in the ipsilateral SVZ and the recruitment of new neuroblasts into the striatum after MCAO and improved survival of new mature neurons. The GDNF receptor GFRalfa1 was up-regulated in the SVZ 1 week after MCAO and was coexpressed with markers of dividing progenitor cells.
[266] Rat Group 1, control = 7; group 2, MSC-EGFP = 6+3; group 3, MSC-GDNF = 7+3; group 4, MSC-BDNF = 7+3; group 5, MSC-CNTF = 7+3; group 6, MSC-NT3 = 7+3 90 min. transient MCAO 5 × 105 adenoviral vector for each EGFP, GDNF, BDNF, CNTF, NT3 genes transfeted-human MSC were intrastriatal injected. After MCAO Rats that received MSC-BDNF or MSC-GDNF showed significantly more functional recovery and reduced ischemic damage on MRI than controls Rat that received MSC-CNTF or MSC-NT3 showed neither functional recovery nor ischemic damage reduction compared to controls.
[336] Rat Control = 6+6+ 4; hMSC = 6+ 6+4; GDNF hMSC = 6+6+4+4+4+4 Normal = 4 Permanent MCAO GDNF-hMSCs were i.v. infused (1.0 × 107) 3 hrs later MCAO Rats receiving GDNF-hMSCs or hMSCs exhibited increased recovery from ischemia compared with the control group, but the effect was greater in the GDNF-hMSC group.
EGF [342] Rat 50 Transient global cerebral ischemia was induced by cardiac arrest, and resuscitation was started at 7 min. 1° xperiement: I.c.v. EGF injection by osmotic minipump (0.5 μl/hr, at 400 or 40 ng/d). Albumin was in 3- and 30-fold molar excess compared with EGF; 2° experiment: EGF (400 ng/d) 1° experiment: 2 days after ischemia for 7 days; 2° experiment: 21 days after ischemia for 7 days EGF mRNA was not detected in either the control or the postischemic rat brain. Heparin-binding EGF-like GF (HB-EGF) mRNA expression was rapidly increased in the CA3 sector and the DG of the hippocampus, cortex, thalamus, and cerebellar granule and Purkinje cell layers. EGF receptor mRNA also showed an increase in the CA3 sector and DG.
[343] Mice 20 min. MCAO I.c.v. EGF and albumin augments 100-fold neuronal replacement in the injured striatum after cerebral ischemia. Newly born immature neurons migrate into the ischemic lesion and differentiate into mature parvalbumin-expressing neurons, replacing more than 20% of the interneurons lost by 13 weeks after ischemia and representing 2% of the total BrdU-labeled cells.
[344] Rat Transient forebrain ischemia bilateral: coagulation of vertebral arteries and bilaterally 6 min. BCCAO. A cocktail of FGF-2 and EGF was infused i.c.v. bilaterally by osmotic minipumps (1.0 μl/hr, resulting in a delivery of 1440 ng of each GF per day, per brain for 3 days) Days 2–5 after ischemia Endogenous progenitors proliferate in response to ischemia and subsequently migrate into the hippocampus to regenerate new neurons. I.c.v. infusion of GFs markedly augments these responses, thereby increasing the number of newborn neurons.
[296] endothelin-1 (ET-1) rat model 22 MCAO via intracerebral microinjection of ET-1 Combined i.c.v. EGF/bFGF (0.48 μg/factor/day) via osmotic pump. 10 min. after ischemia. EGF/bFGF substantially increased the infarct volume in ischemic animals. They induced increase in cell proliferation in the lateral ventricle 14 days after surgery and in the striatum.
[345] rat 80 min. MCAO Either adenovirus-expressing HB-EGF (Ad-HB-EGF) or Ad-LacZ injected into the lateral ventricle on the ischemic side (1.1 × 1010 pfu/ml) 3 days after MCAO There was no significant difference in infarct volume between the 2 groups. Treatment with Ad-HB-EGF significantly increased the number of BrdU+ cells in the SVZ. BrdU+ cells differentiated into mature neurons in the striatum on the ischemic side but seldom the cells given Ad-LacZ. Enhancement of angiogenesis at the peri-infarct striatum was also observed in Ad-HB-EGF–treated rats.
FGF [292] Rat Permanent MCAO bFGF (45 mcg/kg/hr) or vehicle were infused i.v. for 3 hrs. 30 min. after MCAO After 24 hr, neurological deficits and infarct volume were significantly bFGF-treated animals compared to controls. Labeled bFGF crossed the damaged BBB to enter the ischemic (but not the non-ischemic) hemisphere
[293] Rat Permanent MCAO i.v. injection of bFGF, 150 μg/kg, or vehicle. 2 hrs after MCAO Treatment with bFGF showed a significant improvement in functional tests, and a reduction of volume of cortical infarction
[294] Rat Sham-bFGF/sham vehicle = 4–6/6; Stroke-BFGF/stroke vehicle = 5–6/6 MCAO bFGF (0.5 mcg) or vehicle were administered intracisternally 24 and 48 hrs after MCAO Infarct size did not differ among rats with or without bFGF treatment. bFGF increased cell proliferation in the ipsilateral SVZ and DG
[295] Rat Transient MCAO EGF+FGF-2 (each 1.44 ng/d) was continuously administered i.c.v. for 3 days On day 1 of reperfusion GFs infusion enhances post-ischemic progenitor cell proliferation by 5 days of reperfusion and neuronal maturation by 21 days of reperfusion in both the DG and SVZ in the adult rat brain.
[297] Rat 6 for group 2 hrs transient MCAO Non-modified MSCs, FGF-2–modified MSCs with HSV-1 1764/-4/pR19/ssIL2-FGF-2, or PBS were administered intracerebrally. 24 hrs after transient MCAO Animals with FGF-2–modified MSCs after stroke demonstrated significant functional recovery, reduction in infarction volume and higher FGF-2 production in treated brain compared with the other groups.
[298] Rat Transient MCAO A replication defective, recombinant adenovirus vector expressing FGF-2, was injected i.c.v. 2 hrs after MCAO The treatment group showed significant recovery and decreased infarct volume compared with the vehicle-treated groups. FGF-2 gene transfer concentration was increased significantly not only in cerebrospinal fluid but also in cerebral substance in the lesioned and treated animals
BDNF [278] Rat 44 MCAO NGF and BDNF were continuously delivered to the striatum at biologically active levels via recombinant (rAAV) gene Transfer 4–5 weeks prior to 30 MCAO Mild functional improvements 3–5 weeks after the insult, in agreement with a small but significant increase of the survival of dorsolateral striatal neurons.
[270] Rat BDNF n = 12; vehicle n = 13 2-hr MCAO Continuous i.v. infusion of BDNF (300 mg/kg per hr) for 3 hrs 30 min. after occlusion Neurological deficit and infarct volume were improved in BDNF-treated animals versus controls. In these animals Bax-positive neurons were significantly reduced whereas Bcl-2–positive neurons were significantly increased in the penumbra.
[276] (nt4−/−) (bdnf+/−) or wt mice Mice lacking both alleles for neurotrophin-4 (nt4−/−) or deficient in a single allele for brain-derived neurotrophic factor (bdnf+/−) 1-hr MCAO (nt4−/−) and (bdnf+/−) mice exhibited larger cerebral infarcts compared to wild-type Lesions were larger in nt4−/− mice after MCAO
[277] Rat GFPrAAV = 10; BDNF-rAAV = 14 Global forebrain ischemia by BCCAO carotid arteries combined with hypotension Unilateral injections into two sites in the DG hilus of rAAV containing either the GFP gene (GFPrAAV) or the genes for GFP and BDNF (BDNF-rAAV), under control of the rat NSE promoter 5 weeks before ischemia In treated animals BDNF induced a functional response that inhibits the formation of new dentate granule cells triggered by global forebrain ischemia.
[284] Rat Permanent MCAO i.v. injection of 50 μg/rat of either BDNF alone or BDNF as a conjugate with the murine OX26 MAb to the rat TfR After MCAO BDNF resulted in both a reduction in stroke volume and an improvement in functional outcome following delayed i.v. administration in regional brain ischemia, provided the neurotrophin is conjugated to a BBB molecular Trojan horse as BDNF-MAb conjugate.
[282] BDNF+/− mice and wt 40 min. MCAO BDNF+/− mice had a significantly improved motor function compared with wild-type mice. There was no effect of BDNF reduction on infarct volume. Neurogenesis is induced following experimental stroke, and in the striatum of BDNF+/− mice significantly increased numbers of neuroblasts compared with wild-type.
[279] Rat Distal MCAO Authors found permanent reduction of BDNF and its full-length receptor, TrkB, in the infarcted core and a transient increase in BDNF immunoreactivity in the internal region of the border of the infarct, suggesting that BDNF regulates its full-length TrkB receptor in cortical neurons of the penumbra area and prevents their death.

MCAO: middle cerebral artery occlusion; i.v.: intravenously; s.c.: subcutaneously; i.p.: intraperitoneally; BBB: blood brain barrier; SVZ: sub-ventricular zone; DG: dentate grisus; i.c.v.: intracerebroventricularly; NSCs: neural stem cells; BCCAO: bilateral common carotid artery occlusion; SN: substantia nigra.

Brain–derived neurotrophic factor

BDNF, a member of the nerve growth factor (NGF) family, is the most abundantly expressed neurotrophin in the mature CNS. It acts through a high-affinity cell surface receptor (TRKB) [269], supports the survival of many types of neurons and prevents neuronal death after traumatic events like cerebral ischemia by activating intracellular protein kinases B, MAPKs and ERKs [270]. BDNF is also related to synaptic and axonal plasticity associated with memory, learning and sensory-motor improvement [271, 272]. It also increases the number of neurons in newborns and induces neurogenesis [273275]. With regard to these findings, BDNF knockout mice developed larger infarcts, and blockade of endogenous BDNF reduced the survival of neurons after an ischemic insult [276, 277]. Intrastriatal infusion of BDNF in adult rats before ischemia induced increased neuronal survival and improved functional recovery [278].

Ferrer et al. [279], in an ischemia model of MCAO rats, found a permanent reduction of BDNF and its full-length receptor, TRKB in the infarcted core as well a transient increase in BDNF immunoreactivity in the penumbra area 12 hrs after MCA occlusion. The authors proposed a role for BDNF in preventing neuronal death in the penumbra area by up-regulating the full-length TRKB receptor.

In 1997, Schabitz et al. [280] showed that pre-treatment of MCAO rats with intraventricular BDNF reduces infarct size. These results were confirmed by Nomura et al. [267], who demonstrated that intravenous administration of hMSCs expressing the BDNF gene in MCAO rats reduced the infarct volume. These favourable effects were proven only in acute phase stroke, whereas later delivery seems to attenuate intrinsic neurogenerative responses [281]. After transient MCAO in BDNF+/− mice were compared with wild-type mice, BDNF was shown to provide functional improvement by inducing neurogenesis and not by influencing infarct volume. To support these results in BDNF+/− mice, the number of neuroblasts in the striatum was found to be significantly increased [282]. However, since BDNF does not seem to cross the BBB, it can be argued as to whether administration of this factor, when the BBB is not disrupted, is protective in brain ischemia [283, 284]. Thus, the most effective means of administration remains to be assessed.

Current evidence and future perspectives

After transient MCAO, BDNF mRNA is up-regulated in cortical neurons suggesting that BDNF potentially plays a neuroprotective role in focal cerebral ischemia. No clinical studies are available using BDNF in stroke patients. Despite promising results from a pre-clinical study mostly in the acute phase of stroke, further experimental studies should be implemented to assess whether intravenously administered BDNF could be effective in stroke recovery before starting clinical trials.

Basic fibroblast growth factor

bFGF or FGF2 is probably the most potent neurotrophic factor in the FGF superfamily, and has mitogenic, angiogenic and neurotrophic properties [285]. Experimental studies showing that the progenitor cell population in the SVZ is reduced 50% in FGF2 knockout rats suggest that bFGF is involved in neurogenesis [286].

bFGF is widely distributed in neuronal and non-neuronal tissues and has additional effects on mitogenic activity and vasodilation [287, 288]. Preliminary experimental studies showed up-regulated expression of FGF in brain after ischemic injury [289]. Additionally, bFGF is implicated in neurogenesis; after endogenous administration of FGF, cells co localize with neurons and markers of proliferating cells such as BrdU and NeuN [290].

Intravenous administration of high doses of bFGF within hours after stroke onset reduces infarct size (24–50% reduction of infarct volume) in animals subjected to MCAO [291], showing that intravenous bFGF crosses the damaged BBB [292]. These effects were confirmed by Li et al. [293], who showed that infusion of bFGF 2 hrs after stroke onset significantly improved sensorimotor functions and reduced infarct volume. Although the mechanism of neuroprotection by intravenous bFGF is unclear, it is supposed that it is related to a direct cytoprotection of the cells in the penumbra area [293].

In contrast, intracisternal administered bFGF in a rat stroke model does not seem to influence infarct size, but did enhance neuronal sprouting, synapse formation and progenitor cell proliferation and differentiation [291294]. Interestingly, bFGF mRNA expression significantly increases as early as 60 min. following ischemia and remains elevated for up to 2 weeks after onset in a focal cerebral ischemia induced by transient occlusion of the middle cerebral and both common carotid arteries [285]. Moreover, a combined treatment with intraventricularly infused EGF and FGF-2 in adult spontaneously hypertensive rats [295] or in ischemic rats by MCAO occlusion [296] showed that this combined treatment further increases ischemia-induced endogenous neurogenesis and cell maturation. Also, administration of BMSCs engineered to produce FGF2 [297] and recombinant adenovirus vector expressing FGF2 decreased infarct size [298], supporting a neuroprotective role and a neurogenesis-stimulating property for this GF, taking advantage of a less invasive administration method.

Current evidence and future perspectives

The results of experimental studies support the implementation of a phase III clinical trial of bFGF in human beings. However, a North American phase II/III trial was halted after review of data from 300 patients with acute ischemic stroke owing to higher mortality rates in the treated group compared with the control group [299]. The continuing European-Australian phase II/III randomized trial in 286 acute ischemic stroke patients confirmed that intravenous administration of Trafermin (bFGF, 5 or 10 mg) or placebo intravenously infused over 24 hrs did not produce any significant neuroprotection, causing instead dose-dependent hypotension and an increased mortality rate in treated patients [300].

Recently, one study investigated bFGF serum levels in 30 patients with acute cerebral infarction and found that serum bFGF increased significantly after cerebral infarction in comparison with a control group, peaked on day 3, and remained significantly elevated until day 14 after stroke. In this study, it was also found that bFGF levels correlated with infarct size and clinical prognosis [301]. Thus, because of the number of side effects and increased mortality reported in the first clinical studies, further experimental studies are necessary to assess whether it is possible to achieve a pharmacologically significant therapeutic level in brain, minimizing peripheral side effects.

Insulin-like growth factor 1

IGF-1, a peptide produced primarily in the liver and brain, is best characterized as a mediator of growth hormone actions because of its important role in tissue growth regulation. A less-recognized action of serum IGF-1 is its multipotent neurotrophic activity [302]. Circulating IGF-1 can cross the BBB [303306] and modulates brain function in many different ways [306308]. It is a well known angiogenic factor [309] that also plays an essential role in normal growth and brain development [310], other than in modulation of vessel formation during brain development [311], and possibly in age-related changes in the brain vasculature [312]. It was also reported to enhance endothelial function through its anti-inflammatory and anti-apoptotic properties [313].

In past years, it was supposed that both brain-derived and circulating IGF-1 acted as a neuroprotective signal against acute ischemic brain injury [314, 315]. Actually, the role of IGF-1 in ischemic stroke is controversial, even in consideration of the wide variability of serum IGF-1 levels among individuals [316]. For instance, serum IGF-1 levels in human beings may be depressed after acute stroke [314, 315], whereas increased IGF-1 levels have been detected proximal to ischemic lesions in rodent models [317], suggesting a protective role of IGF-1 [302]. Many later studies showed that IGF-1 administration protects adult brain from experimental ischemia [318320]. Intranasal (IN) delivery of rhIGF-I in a MCAO rat model significantly reduced the infarct volume versus controls, and improved all neurologic deficits [321].

In permanent distal MCAO in mice, long-term cerebral IGF-1 overexpression by the AAV transduction system through stereotaxic injection significantly improved motor performance, reduced the volume of cerebral infarction, and increased neovessel formation in the peri-infarct and injection needle tract area. Increased vascular density was associated with increased local vascular perfusion.

AAV-IGF-1 treatment was also observed to enhance neurogenesis in the SVZ [322]. Moreover, intravenous infusion of IGF-1 significantly decreased both lesion volume and apoptosis in diabetic animals after MCAO [323]. Therefore, serum IGF-1 would be expected to improve stroke outcome.

Since Schwab et al. observed decreased plasma levels of IGF-I and its receptor, IGFBP-3 in patients with acute stroke over a 10-period after onset of symptoms [314], the pathophysiological mechanism and the time course of IGF-1 levels after ischemic stroke must be better clarified.

Current evidence and future perspectives

To date, no clinical trials have been implemented to assess the benefit of IGF-1 on recovery in stroke patients. However, while the experimental studies to date are promising, a cautious approach to clinical application is needed since it is not clear how IGF-1 exerts its neuroprotective role.

Glial cell line-derived neurotrophic factor

Glial cell line-derived neurotrophic factor (GDNF) is a potent neurotrophic factor, acting mostly through the extracellular glycosylphospatidylinositol (GPI)-linked receptor, GFR-1 and the transmembrane tyrosine kinase, c-Ret. It was purified and characterized in 1993 as a GF promoting survival and differentiation of embryonic midbrain dopaminergic neurons [324, 325]. Subsequently, GDNF was shown to be a very potent trophic factor for spinal motoneurons [326] and central noradrenergic neurons [327], and to promote survival and differentiation of many peripheral neurons such as sympathetic, parasympathetic, sensory and enteric neurons [328]. Therefore, this trophic factor raised the expectation that it might have potential as a therapeutic agent for the treatment of neurodegenerative diseases.

In MCAO rats, topical application of GDNF reduced both infarct size and brain oedema [329331]. The authors suggested that the ameliorative effect of GDNF is related to its anti-apoptotic properties [329]. GDNF receptor expression was up-regulated in the penumbral area and GDNF delivery seemed to improve short-term neuronal survival in MCAO rats [332, 333]. Several findings suggest that GDNF may affect neurogenesis after stroke. Exogenous infusions of GDNF increased focal ischemia-induced cell proliferation of the ipsilateral DG of the subgranular zone (SGZ) in adult rats [334]. More recently, Kobayashi et al. [335] infused GDNF intra-striatally into rats after a 2-hr MCAO to clarify whether GDNF delivery during the first week after MCAO affects cell proliferation in the SVZ and recruits neuroblasts into the damaged striatum. The results of this study showed that GDNF infusion promoted survival of new neurons during the post-ischemic phase without influencing the speed of neuronal maturation. Since GFRα1 levels were also elevated in the SVZ one week after stroke, the authors suggested that the effects of GDNF on cell proliferation are mediated by receptors present on stem cells.

Intravenously administered GDNF-gene modified hMSCs in MCAO rat models reduced the infarct volume, improved behavioural performance, and increased GDNF levels in the infarcted hemisphere [266, 336]. Intrastriatal injections of recombinant lentivirus (rLV) or adenovirus (rAAV) carrying the GDNF gene (rLV-GDNF or rAAV-GDNF) were performed in rats after transient MCAO. Both routes of GDNF gene delivery effectively promoted high striatal GDNF levels, which persisted several weeks after stroke. In contrast with previous experiments, the treatment did not increase neuron survival or improve functional recovery. The authors proposed that the effect of GDNF is complex and may depend on several factors, primarily protein levels, degree and pattern of receptor expression on cells in the damaged areas, route of delivery, type of cells transduced, and GDNF’s actions on neuronal transmission at the site of delivery [333].

Current evidence and future perspectives

Despite pre-clinical studies demonstrating that GDNF improves neurogenesis and cell survival and differentiation, these effects are controversial. Thus, it is too early to test this neurotrophin in clinical studies, and more experimental studies should be performed to explain the heterogeneous results from previous studies.

Heparin-binding EGF-like growth factor

Heparin-binding EGF-like growth factor (HB-EGF) is a member of the EGF family, originally described as a macrophage-derived mitogenic and chemotactic factor of vascular smooth muscle cells [337]. HB-EGF is synthesized in a membrane-anchored form (proHB-EGF) and released as a secreted form, soluble HB-EGF [338]. HB-EGF is widely distributed in neurons and neuroglia throughout rat brain [339]. A number of studies demonstrated its neurotrophic activities in vitro, such as survival-promoting and neurite outgrowth effects [340], and neuroprotective effects against anoxia and nitric oxide toxicity [341]. In vivo, both HB-EGF and EGFR mRNAs were up-regulated following ischemia in rats, mostly in the hippocampal CA3 sector and the DG [342]. Teramoto et al. [343] first demonstrated that intraventricular administration of EGF amplifies proliferation of neural progenitors in the SVZ and the replacement of striatal interneurons after MCAO. Nakatomi et al. (2002) showed that a combined treatment with EGF and bFGF increased the number of NeuN+ neurons 4 weeks after global ischemia in rats, confirming the role of this neurotrophin in neurogenesis [344]. This finding was recently confirmed by Baldauf et al. [296] who found that EGF/bGF infusion into the lateral ventricle in a model of MCAO rats, induced by endothelin injection, influences neurogenesis and cell proliferation in the striatum, supporting the notion that administration of these GFs accelerates cell migration from the ventricle and enhances proliferation [296]. Surprisingly, chronic administration of EGF/bFGF results in an enlarged infarct volume, raising some doubts about the treatment schedule and time of neurotrophin administration [345]. Watanabe injected adenovirus transferring the HB-EGF gene into the lateral ventricle on the ischemic side in a transient cerebral ischemia rat model and observed not only increased neurogenesis, but also enhanced angiogenesis in the peri-infarct striatum on day 8 in HB-EGF-treated rats, improving neurological functional recovery [298]. The proposed hypothesis was that HB-EGF promotes angiogenesis by inducing VEGF secretion from vascular smooth muscle cells, which enhances proliferation of endothelial cells.

Current evidence and future perspectives

Experimental models show that post-ischemic EGF/bGF intraventricular administration and gene therapy using the HB-EGF gene improves neurogenesis and angiogenesis. However, there are doubts about treatment schedules and the appropriate time of administration. Additional pre-clinical studies are necessary to better ascertain dosage, timing and the best method of administration.

Discussion

Post-stroke brain remodelling proceeds through several different mechanisms including neurogenesis, angiogenesis, axonal plasticity and white matter changes. Post-stroke neurogenesis and angiogenesis consists of different steps as well. Endogenous neurogenesis in response to brain ischemia requires neuronal stem cells and progenitor cells proliferation, differentiation of neuronal progenitor cells and migration of neuroblasts to the peri-ischemic area where they integrate into the parenchymal tissue as mature neurons. Angiogenic responses to pathophysiological conditions include endothelial cells proliferation and migration, tube formation and branching and anastomosis [346]. Our increasing understanding of the complex mechanism underlying endogenous responses to brain ischemia may identify therapeutic options that will promote post-stroke brain recovery.

GFs are normally expressed in adult brain [108, 155] and are up-regulated in response to brain ischemia [36, 40, 110, 113, 184, 242, 246, 247]. Moreover, knockout mouse studies confirm that deficiencies in select GFs impairs the response to brain ischemia, leading to a larger infarct size and poorer clinical outcome [41, 91].

Haematopoietic GFs, through their pleiotropic effects, play a significant role in enhancing ischemic brain remodelling at different levels, and counteract the ischemia-induced phenomena that contributes to brain damage after stroke, such as inflammation and oedema.

G-CSF plays a significant role in post-stroke neurogenesis by mobilizing endogenous CD34+ stem cells and promoting their homing to the site of injury [49, 50] and in promoting cell proliferation from the SGZ of DG [6568] as well as reducing post-stroke brain damage. A significant reduction in brain oedema and BBB damage after G-CSF administration is probably mediated by the suppression of injury-induced pro-inflammatory responses and T-cell infiltration [4547]. EPO promotes post-ischemic brain remodelling, influencing stem cell differentiation and playing an angiogenic role [8891, 129, 131]. Additionally, EPO regulates the expression of genes involved in apoptosis [115118], to reduce the production of pro-inflammatory cytokines [120] and brain oedema [123].

GM-CSF results in with improved brain hemodynamic parameters [161], favours proliferation and structural changes in intraparenchymal vessels after stroke [158, 163], enhances leptomeningeal collateral growth [163] and counteracts metabolic damage subsequent to hypotension [162]. In addition to this, GM-CSF administration results in a positive modulation of apoptosis-related genes leading to a significant decrease in neuronal apoptosis [155, 160]. VEGF and its receptor (VEGFR-2) play a significant role in the initial phase of angiogenesis. Angiopoietins are involved in the maturation, stabilization and remodelling of vessels, and VEGF overexpression resulted in a higher capillary density but not a corresponding increase in blood flow [199]. Moreover, VEGF counteracted apoptosis and reduced brain oedema.

Taken together, the results of pre-clinical studies show that bEGF, VEGF, G-CSF, EGF, FGF-2, IGF and GDNF [343, 334] are up-regulated after ischemia [294, 317, 347350] and stimulate both in vitro and in vivo neurogenesis and angiogenesis. In particular, in experimental models, they were shown to contribute to infarct size and brain oedema reduction, cell proliferation, and improved survival of new mature neurons, apoptosis reduction, increased neovessel formation and improved clinical outcome (Tables 1 and 2). bFGF, VEGF, EPO and G-CSF have all shown potential effectiveness in stroke recovery in experimental models.

On the basis of these promising results, some of these factors were tested in clinical trials. Completed EPO trials [138, 139] involving patients with hyperacute stroke (treatment within 8 hrs of symptom onset) reported a positive, but insignificant [138] tendency, both in terms of infarct size reduction and clinical outcome, suggesting a possible neuroprotective effect in the treatment arm. The results from three G-CSF trials in stroke patients showed that it is safe, well-tolerated, and that its administration results in mobilization of mobilizing bone marrow CD34+ stem cells [69]. Despite this, the results on clinical outcome from two clinical trials involving patients with recent stroke were uncertain suggesting a positive, but non-significant effect on 90-day disability (BI) [69], neurologic function (NIHSS) after 20 days [71] and long-term outcome (12-month assessment) [70].

bFGF administration resulted in dose-dependent hypotension and increased the mortality rate in treated patients compared with the control group [300].

As a consequence of the methodological heterogeneity between different trials and the small patient populations, there is a substantial lack of evidence for the administration of GFs to patients with stroke [72]. Thus, although data from the existing trials suggest that EPO and G-CSF are safe and well-tolerated and may have a positive effect in terms of neuroprotection (reduced infarct size after EPO administration) and neuronal repair (G-CSF), further studies are necessary to assess the safety and efficacy of GF use in stroke patients.

The failure of clinical trials with bFGF raises questions about the importance of the BBB in achieving a pharmacologically significant therapeutic level of GFs in brain, and minimizing side effects in the peripheral tissues. For instance, intranasal delivery may be considered as a practical, non-invasive method of bypassing the BBB to deliver therapeutic agents to the brain [351]. Avoiding side effects in peripheral tissues is obviously an important issue. Additionally, the development of structural and functional variants of EPO with reduced or absent haematopoietic functions represents a challenge in stroke treatment [139].

The optimal timing of administration of GFs is still unclear, since most published studies on stroke patients focus on recent stroke (enrolment from 7 to 30 days from stroke onset), and data on their use in acute and chronic stroke are lacking. For instance, Shyu et al. [70] reported a greater clinical improvement in the subgroup (n = 4) of patients who received G-CSF within 24 hrs, but this finding should be confirmed. Also, the safety of GFs administered in this subgroup of patients should be assessed. With regard to VEGF, it was more effective over 48 hrs, so as to avoid the increased risk of brain oedema in the acute phase [200, 203]. However, IGF-1 was not beneficial in the acute phase [302]. With regard to G-CSF, a few pre-clinical studies [261, 352] suggest a possible positive effect in chronic stroke.

The current uncertainty regarding the use of GFs as stroke treatments derives from the fact that none of the trials definitively addressed the mechanisms by which GFs might work after stroke. Focusing on the potential mechanisms of action determined in pre-clinical studies will help investigators decide when to administer treatment relative to stroke onset.

References

  • 1.Bliss T, Guzman R, Daadi M, et al. Cell transplantation therapy for stroke. Stroke. 2007;38:817–26. doi: 10.1161/01.STR.0000247888.25985.62. [DOI] [PubMed] [Google Scholar]
  • 2.Locatelli F, Bersano A, Ballabio E, et al. Stem cell therapy in stroke. Cell Mol Life Sci. 2009;66:757–72. doi: 10.1007/s00018-008-8346-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Kaplan MS, Bell DH. Mitotic neuroblasts in the 9-day-old and 11-month-old rodent hippocampus. J Neurosci. 1984;4:1429–41. doi: 10.1523/JNEUROSCI.04-06-01429.1984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Kuhn HG, Dickinson-Anson H, Gage FH. Neurogenesis in the dentate gyrus of the adult rat: age-related decrease of neuronal progenitor proliferation. J Neurosci. 1996;16:2027–33. doi: 10.1523/JNEUROSCI.16-06-02027.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Luskin MB, Zigova T, Soteres BJ, et al. Neuronal progenitor cells derived from the anterior subventricular zone of the neonatal rat forebrain continue to proliferate in vitro and express a neuronal phenotype. Mol Cell Neurosci. 1997;8:351–66. doi: 10.1006/mcne.1996.0592. [DOI] [PubMed] [Google Scholar]
  • 6.Alvarez-Buylla A, Garcia-Verdugo JM. Neurogenesis in adult subventricular zone. J Neurosci. 2002;22:629–34. doi: 10.1523/JNEUROSCI.22-03-00629.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Sanai N, Tramontin AD, Quiñones-Hinojosa A, et al. Unique astrocyte ribbon in adult human brain contains neural stem cells but lacks chain migration. Nature. 2004;427:740–4. doi: 10.1038/nature02301. [DOI] [PubMed] [Google Scholar]
  • 8.Curtis MA, Eriksson PS, Faull RL. Progenitor cells and adult neurogenesis in neurodegenerative diseases and injuries of the basal ganglia. Clin Exp Pharmacol Physiol. 2007;34:528–32. doi: 10.1111/j.1440-1681.2007.04609.x. [DOI] [PubMed] [Google Scholar]
  • 9.Sprigg N, Bath P. Pharmacological enhancement of recovery from stroke. Stroke Review. 2005;8:33–40. [Google Scholar]
  • 10.Ransome MI, Turnley AM. Systemically delivered erythropoietin transiently enhances adult hippocampal neurogenesis. Neurochem. 2007;102:1953–65. doi: 10.1111/j.1471-4159.2007.04684.x. [DOI] [PubMed] [Google Scholar]
  • 11.Kalluri HS, Vemuganti R, Dempsey RJ. Lack of response to epidermal growth factor in adult neural progenitor cells. Neuroreport. 2005;16:835–8. doi: 10.1097/00001756-200505310-00011. [DOI] [PubMed] [Google Scholar]
  • 12.Burrows RC, Wancio D, Levitt P, et al. Response diversity and the timing of progenitor cell maturation are regulated by developmental changes in EGFR expression in the cortex. Neuron. 1997;19:251–67. doi: 10.1016/s0896-6273(00)80937-x. [DOI] [PubMed] [Google Scholar]
  • 13.Le Beau M, Lemons R, Carrino J, et al. Chromosomal localization of the human G-CSF gene to 17q11 proximal to the breakpoint of the t(15; 17) in acute promyelocytic leukemia. Leukemia. 1987;1:795–9. [PubMed] [Google Scholar]
  • 14.Nagata S, Tsuchiya M, Asano S, et al. Molecular cloning and expression of cDNA for human granulocyte colony-stimulating factor. Nature. 1986;319:415–8. doi: 10.1038/319415a0. [DOI] [PubMed] [Google Scholar]
  • 15.Schneider A, Kuhn HG, Schäbitz WR. A role for G-CSF (granulocyte-colony stimulating factor) in the central nervous system. Cell Cycle. 2005;4:1753–7. doi: 10.4161/cc.4.12.2213. [DOI] [PubMed] [Google Scholar]
  • 16.Hareng L, Hartung T. Induction and regulation of endogenous granulocyte colony-stimulating factor formation. Biol Chem. 2002;383:1501–17. doi: 10.1515/BC.2002.172. [DOI] [PubMed] [Google Scholar]
  • 17.Kaushansky K. Lineage-specific hematopoietic growth factors. N Engl J Med. 2006;354:2034–45. doi: 10.1056/NEJMra052706. [DOI] [PubMed] [Google Scholar]
  • 18.Koeffler HP, Gasson J, Ranyard J, et al. Recombinant human TNF alpha stimulates production of granulocyte colony-stimulating factor. Blood. 1987;70:55–9. [PubMed] [Google Scholar]
  • 19.Kaushansky K, Lin N, Adamson JW. Interleukin 1 stimulates fibroblasts to synthesize granulocyte-macrophage and granulocyte colony-stimulating factors. Mechanism for the hematopoietic response to inflammation. J Clin Invest. 1988;81:92–7. doi: 10.1172/JCI113316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Vellenga E, Rambaldi A, Ernst TJ, et al. Independent regulation of M-CSF and G-CSF gene expression in human monocytes. Blood. 1988;71:1529–32. [PubMed] [Google Scholar]
  • 21.Wieser M, Bonifer R, Oster W, et al. Interleukin-4 induces secretion of CSF for granulocytes and CSF for macrophages by peripheral blood monocytes. Blood. 1989;73:1105–8. [PubMed] [Google Scholar]
  • 22.Oster W, Lindemann A, Mertelsmann R, et al. Production of macrophage-, granulocyte-, granulocyte-macrophage- and multi-colony-stimulating factor by peripheral blood cells. Eur J Immunol. 1989;19:543–7. doi: 10.1002/eji.1830190320. [DOI] [PubMed] [Google Scholar]
  • 23.Fukunaga R, Ishizaka-Ikeda E, et al. Functional domains of the granulocyte colonystimulating factor receptor. EMBO J. 1991;10:2855–65. doi: 10.1002/j.1460-2075.1991.tb07835.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Hanazono Y, Hosoi T, Kuwaki T, et al. Structural analysis of the receptors for granulocyte colony-stimulating factor on neutrophils. Exp Hematol. 1990;18:1097–103. [PubMed] [Google Scholar]
  • 25.Shimoda K, Okamura S, Harada N, et al. Identification of a functional receptor for granulocyte colony-stimulating factor on platelets. J Clin Invest. 1993;91:1310–3. doi: 10.1172/JCI116330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Morikawa K, Morikawa S, Nakamura M, et al. Characterization of granulocyte colonystimulating factor receptor expressed on human lymphocytes. Br J Haematol. 2002;118:296–304. doi: 10.1046/j.1365-2141.2002.03574.x. [DOI] [PubMed] [Google Scholar]
  • 27.Boneberg EM, Hareng L, Gantner F, et al. Human monocytes express functional receptors for granulocyte colony-stimulating factor that mediate suppression of monokines and interferon-gamma. Blood. 2000;95:270–6. [PubMed] [Google Scholar]
  • 28.Bussolino F, Wang JM, Defilippi P, et al. Granulocyte- and granulocyte-macrophagecolony stimulating factors induce human endothelial cells to migrate and proliferate. Nature. 1989;337:471–3. doi: 10.1038/337471a0. [DOI] [PubMed] [Google Scholar]
  • 29.Shimoda K, Feng J, Muratami H, et al. Jak1 plays an essential role for receptor phosphorylation and Stat activation in response to granulocyte colonystimulating factor. Blood. 1997;90:597–604. [PubMed] [Google Scholar]
  • 30.Tian SS, Lamb P, Seidel HM, et al. Rapid activation of the STAT3 transcription factor by granulocyte colony-stimulating factor. Blood. 1994;84:1760–4. [PubMed] [Google Scholar]
  • 31.Hunter MG, Avalos BR. Phosphatidylinositol 3’- kinase and SH2-containing inositol phosphatase (SHIP) are recruited by distinct positive and negative growthregulatory domains in the granulocyte colony-stimulating factor receptor. J Immunol. 1998;160:4979–87. [PubMed] [Google Scholar]
  • 32.Dong F, Larner AC. Activation of Akt kinase by granulocyte colony-stimulating factor (G-CSF): evidence for the role of a tyrosine kinase activity distinct from the Janus kinases. Blood. 2000;95:1656–62. [PubMed] [Google Scholar]
  • 33.Ward AC, Loeb DM, Soede-Bobok AA, et al. Regulation of granulopoiesis by transcription factors and cytokine signals. Leukemia. 2000;14:973–90. doi: 10.1038/sj.leu.2401808. [DOI] [PubMed] [Google Scholar]
  • 34.To LB, Haylock DN, Simmons PJ, et al. The biology and clinical uses of blood stem cells. Blood. 1997;89:2233–58. [PubMed] [Google Scholar]
  • 35.Han JL, Blank T, Schwab S, et al. Inhibited glutamate release by granulocyte-colony stimulating factor after experimental stroke. Neurosci Lett. 2008;432:167–9. doi: 10.1016/j.neulet.2007.07.056. [DOI] [PubMed] [Google Scholar]
  • 36.Schneider A, Krüger C, Steigleder T, et al. The hematopoietic factor G-CSF is a neuronal ligand that counteracts programmed cell death and drives neurogenesis. J Clin Invest. 2005;115:2083–98. doi: 10.1172/JCI23559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Solaroglu I, Jadhav V, Zhang JH. Neuroprotective effect of granulocyte-colony stimulating factor. Front Biosci. 2007;12:712–24. doi: 10.2741/2095. [DOI] [PubMed] [Google Scholar]
  • 38.Komine-Kobayashi M, Zhang N, Liu M, et al. Neuroprotective effect of recombinant human granulocyte colony-stimulating factor in transient focal ischemia of mice. J Cereb Blood Flow Metab. 2006;26:402–13. doi: 10.1038/sj.jcbfm.9600195. [DOI] [PubMed] [Google Scholar]
  • 39.Solaroglu I, Tsubokawa T, Cahill J, et al. Anti-apoptotic effect of granulocyte-colony stimulating factor after focal cerebral ischemia in the rat. Neurosci. 2006;143:965–74. doi: 10.1016/j.neuroscience.2006.09.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Hasselblatt M, Jeibmann A, Riesmeier B, et al. Granulocyte-colony stimulating factor (G-CSF) and G-CSF receptor expression in human ischemic stroke. Acta Neuropathol. 2007;113:45–51. doi: 10.1007/s00401-006-0152-y. [DOI] [PubMed] [Google Scholar]
  • 41.Sevimli S, Diederich K, Strecker JK, et al. Endogenous brain protection by granulocyte-colony stimulating factor after ischemic stroke. Exp Neurol. 2009;217:328–35. doi: 10.1016/j.expneurol.2009.03.018. [DOI] [PubMed] [Google Scholar]
  • 42.Mun-Bryce S, Rosenberg GA. Matrix metalloproteinases in cerebrovascular disease. J Cereb Blood Flow Metab. 1998;18:1163–72. doi: 10.1097/00004647-199811000-00001. [DOI] [PubMed] [Google Scholar]
  • 43.Hamann GF, Okada Y, Fitridge R, et al. Microvascular basal lamina antigens disappear during cerebral ischemia and reperfusion. Stroke. 1995;26:2120–6. doi: 10.1161/01.str.26.11.2120. [DOI] [PubMed] [Google Scholar]
  • 44.Unemori EN, Bouhana KS, Werb Z. Vectorial secretion of extracellular matrix proteins, matrix-degrading proteinases, and tissue inhibitor of metalloproteinases by endothelial cells. J Biol Chem. 1990;265:445–51. [PubMed] [Google Scholar]
  • 45.Gibson CL, Jones NC, Prior MJ, et al. G-CSF suppresses edema formation and reduces interleukin-1beta expression after cerebral ischemia in mice. J Neuropathol Exp Neurol. 2005;64:763–9. doi: 10.1097/01.jnen.0000179196.10032.dd. [DOI] [PubMed] [Google Scholar]
  • 46.Gorgen I, Hartung T, Leist M, et al. Granulocyte colony-stimulating factor treatment protects rodents against lipopolysaccharide-induced toxicity via suppression of systemic tumor necrosis factor-alpha. J Immunol. 1992;149:918–24. [PubMed] [Google Scholar]
  • 47.Zavala F, Abad S, Ezine S, et al. G-CSF therapy of ongoing experimental allergic encephalomyelitis via chemokine- and cytokine-based immune deviation. J Immunol. 2002;168:2011–9. doi: 10.4049/jimmunol.168.4.2011. [DOI] [PubMed] [Google Scholar]
  • 48.Hartung T, Docke WD, Gantner F, et al. Effect of granulocyte colony-stimulating factor treatment on ex vivo blood cytokine response in human volunteers. Blood. 1995;85:2482–9. [PubMed] [Google Scholar]
  • 49.Jendelova P, Herynek V, Urdzikova L, et al. Magnetic resonance tracking of human CD34+ progenitor cells separated by means of immunomagnetic selection and transplanted into injured rat brain. Cell Transplant. 2005;14:173–82. doi: 10.3727/000000005783983124. [DOI] [PubMed] [Google Scholar]
  • 50.Sykova E, Jendelova P. Magnetic resonance tracking of implanted adult and embryonic stemcells in injured brain and spinal cord. Ann N Y Acad Sci. 2005;1049:146–60. doi: 10.1196/annals.1334.014. [DOI] [PubMed] [Google Scholar]
  • 51.Schäbitz WR, Kollmar R, Schwaninger M, et al. Neuroprotective effect of granulocyte colony-stimulating factor after focal cerebral ischemia. Stroke. 2003;34:745–51. doi: 10.1161/01.STR.0000057814.70180.17. [DOI] [PubMed] [Google Scholar]
  • 52.Six I, Gasan G, Mura E, et al. Beneficial effect of pharmacological mobilization of bone marrow in experimental cerebral ischemia. Eur J Pharm. 2003;458:327–8. doi: 10.1016/s0014-2999(02)02785-1. [DOI] [PubMed] [Google Scholar]
  • 53.Gibson CL, Bath PMW, Murphy SP. G-CSF reduces infarct volume and improves functional outcome after transient focal cerebral ischemia in mice. J Cereb Blood Flow Metab. 2005;25:431–9. doi: 10.1038/sj.jcbfm.9600033. [DOI] [PubMed] [Google Scholar]
  • 54.Schneider A, Wysocki R, Pitzer C, et al. An extended window of opportunity for G-CSF treatment in cerebral ischemia. BMC Biol. 2006;4:36. doi: 10.1186/1741-7007-4-36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Yanqing Z, Yu-Min L, Jian Q, et al. Fibronectin and neuroprotective effect of granulocyte colony-stimulating factor in focal cerebral ischemia. Brain Res. 2006;1098:161–9. doi: 10.1016/j.brainres.2006.02.140. [DOI] [PubMed] [Google Scholar]
  • 56.Minnerup J, Heidrich J, Wellmann J, et al. Meta-analysis of the efficacy of granulocyte-colony stimulating factor in animal models of focal cerebral ischemia. Stroke. 2008;39:1855–61. doi: 10.1161/STROKEAHA.107.506816. [DOI] [PubMed] [Google Scholar]
  • 57.England TJ, Gibson CL, Bath PM. Granulocyte-colony stimulating factor in experimental stroke and its effects on infarct size and functional outcome: a systematic review. Brain Res Rev. 2009;62:71–82. doi: 10.1016/j.brainresrev.2009.09.002. [DOI] [PubMed] [Google Scholar]
  • 58.Lan X, Qu H, Yao W, et al. Granulocyte-colony stimulating factor inhibits neuronal apoptosis in a rat model of diabetic cerebral ischemia. Tohoku J Exp Med. 2008;216:117–26. doi: 10.1620/tjem.216.117. [DOI] [PubMed] [Google Scholar]
  • 59.Kawada H, Takizawa S, Takanashi T, et al. Administration of hematopoietic cytokines in the subacute phase after cerebral infarction is effective for functional recovery facilitating proliferation of intrinsic neural stem/progenitor cells and transition of bone marrow-derived neuronal cells. Circulation. 2006;113:701–10. doi: 10.1161/CIRCULATIONAHA.105.563668. [DOI] [PubMed] [Google Scholar]
  • 60.Sehara Y, Hayashi T, Deguchi K, et al. Decreased focal inflammatory response by G-CSF may improve stroke outcome after transient middle cerebral artery occlusion in rats. J Neurosci Res. 2007a;85:2167–74. doi: 10.1002/jnr.21341. [DOI] [PubMed] [Google Scholar]
  • 61.Morita Y, Takizawa S, Kamiguchi H, et al. Administration of hematopoietic cytokines increases the expression of anti-inflammatory cytokine (IL-10) mRNA in the subacute phase after stroke. Neurosci Res. 2007;58:356–60. doi: 10.1016/j.neures.2007.04.006. [DOI] [PubMed] [Google Scholar]
  • 62.Taguchi A, Wen Z, Myojin K, et al. Granulocyte colony-stimulating factor has a negative effect on stroke outcome in a murine model. Eur J Neurosci. 2007;26:126–33. doi: 10.1111/j.1460-9568.2007.05640.x. [DOI] [PubMed] [Google Scholar]
  • 63.Lee ST, Chu K, Jung KH, et al. Granulocyte colony-stimulating factor enhances angiogenesis after focal cerebral ischemia. Brain Res. 2005;1058:120–8. doi: 10.1016/j.brainres.2005.07.076. [DOI] [PubMed] [Google Scholar]
  • 64.Sehara Y, Hayashi T, Deguchi K, et al. G-CSF enhances stem cell proliferation in rat hippocampus after transient middle cerebral artery occlusion. Neurosci Lett. 2007b;418:248–52. doi: 10.1016/j.neulet.2007.03.047. [DOI] [PubMed] [Google Scholar]
  • 65.Sehara Y, Hayashi T, Deguchi K, et al. Potentiation of neurogenesis and angiogenesis by G-CSF after focal cerebral ischemia in rats. Brain Res. 2007c;1151:142–9. doi: 10.1016/j.brainres.2007.01.149. [DOI] [PubMed] [Google Scholar]
  • 66.Toth ZE, Leker RR, Shahar T, et al. The combination of granulocyte colony-stimulating factor and stem cell factor significantly increases the number of bone marrow-derived endothelial cells in brains of mice following cerebral ischemia. Blood. 2008;111:5544–52. doi: 10.1182/blood-2007-10-119073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Hokari M, Kuroda S, Chiba Y, et al. Synergistic effects of granulocyte-colony stimulating factor on bone marrow stromal cell transplantation for mice cerebral infarct. Cytokine. 2009;46:260–6. doi: 10.1016/j.cyto.2009.02.008. [DOI] [PubMed] [Google Scholar]
  • 68.Shyu WC, Lin SZ, Yang HI, et al. Functional recovery of stroke rats induced by granulocyte colony-stimulating factor–stimulated stem cells. Circulation. 2004;110:1847–54. doi: 10.1161/01.CIR.0000142616.07367.66. [DOI] [PubMed] [Google Scholar]
  • 69.Sprigg N, Bath PM, Zhao L, et al. Granulocyte-colony-stimulating factor mobilizes bone marrow stem cells in patients with subacute ischemic stroke: the Stem cell Trial of recovery EnhanceMent after Stroke (STEMS) pilot randomized, controlled trial (ISRCTN 16784092) Stroke. 2006;37:2979–83. doi: 10.1161/01.STR.0000248763.49831.c3. [DOI] [PubMed] [Google Scholar]
  • 70.Shyu WC, Lin SZ, Lee CC, et al. Granulocyte colony-stimulating factor for acute ischemic stroke: a randomized controlled trial. CMAJ. 2006;174:927–33. doi: 10.1503/cmaj.051322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Zhang JJ, Deng M, Zhang Y, et al. A shortterm assessment of recombinant human granulocyte-stimulating factor (RHG-CSF) in treatment of acute cerebral infarction. Cerebrovascular Diseases. 2006;21:143. [Google Scholar]
  • 72.Bath PMW, Sprigg N. Colony stimulating factors (including erythropoietin, granulocyte colony stimulating factor and analogues) for stroke. Cochrane Database Syst Rev. 2007 doi: 10.1002/14651858.CD005207.pub3. Issue 2. Art. No.: CD005207. DOI: 10.1002/14651858.CD005207.pub3. [DOI] [PubMed] [Google Scholar]
  • 73.Minnerup J, Schäbitz WR. Multifunctional actions of approved and candidate stroke drugs. Neurotherapeutics. 2009;6:43–52. doi: 10.1016/j.nurt.2008.10.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Schäbitz WR, Laage R, Schwab S, et al. AX 200 (G-CSF). Abstracts. 17th European Stroke Conference, Nice, France, May 13–16, 2008. Cerebrovas Dis. 2008:62. doi: 10.1159/000132086. [DOI] [PubMed] [Google Scholar]
  • 75.Schäbitz WR, Schneider A. New targets for established proteins: exploring G-CSF for the treatment of stroke. Trends Pharmacol Sci. 2007;28:157–61. doi: 10.1016/j.tips.2007.02.007. [DOI] [PubMed] [Google Scholar]
  • 76.Ebert BL, Bunn HF. Regulation of the erythropoietin gene. Blood. 1999;94:1864–77. [PubMed] [Google Scholar]
  • 77.Jacobs K, Shoemaker C, Rudersdorf R, et al. Isolation and characterization of genomic and cDNA clones of human erythropoietin. Nature. 1985;313:806. doi: 10.1038/313806a0. [DOI] [PubMed] [Google Scholar]
  • 78.Rankin EB, Biju MP, Liu Q, et al. Hypoxia-inducible factor–2 (HIF-2) regulates hepatic erythropoietin in vivo. J Clin Invest. 2007;117:1068–77. doi: 10.1172/JCI30117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Ehrenreich H, Aust C, Krampe H, et al. Erythropoietin: novel approaches to neuroprotection in human brain disease. Metab Brain Dis. 2004;19:195–206. doi: 10.1023/b:mebr.0000043969.96895.3c. [DOI] [PubMed] [Google Scholar]
  • 80.Zhande R, Karsan A. Erythropoietin promotes survival of primary human endothelial cells through PI3K-dependent, NF-kB-independent upregulation of Bcl-xL. Am J Physiol Heart Circ Physio. 2007;292:2467–74. doi: 10.1152/ajpheart.00649.2006. [DOI] [PubMed] [Google Scholar]
  • 81.Zanjani ED, Poster J, Burlington H, et al. Liver as the primary site of erythropoietin formation in the fetus. J. Lab. Clinical. Med. 1977;89:640–4. [PubMed] [Google Scholar]
  • 82.Dame C, Kirschner KM, Bartz KV, et al. Wilms tumor suppressor, Wt1, is a transcriptional activator of the erythropoietin gene. Blood. 2006;107:4282–90. doi: 10.1182/blood-2005-07-2889. [DOI] [PubMed] [Google Scholar]
  • 83.Fisher JW. Erythropoietin: physiologic and pharmacologic aspects. Proc Soc Exp Biol Med. 1997;216:358–69. doi: 10.3181/00379727-216-44183. [DOI] [PubMed] [Google Scholar]
  • 84.Marti HH, Gassmann M, Wenger RH, et al. Detection of erythropoietin in human liquor: intrinsic erythropoietin production in the brain. Kidney International. 1997;51:416–8. doi: 10.1038/ki.1997.55. [DOI] [PubMed] [Google Scholar]
  • 85.Bernaudin M, Marti HH, Roussel S, et al. A potential role for erythropoietin in focal permanent cerebral ischemia in mice. J Cereb Blood Flow Metab. 1999;19:643–51. doi: 10.1097/00004647-199906000-00007. [DOI] [PubMed] [Google Scholar]
  • 86.Vogt C, Pentz S, Rich IN. A role for the macrophage in normal hemopoiesis: III. In vitro and in vivo erythropoietin gene expression in macrophages detected by in situ hybridization. Exp Hematol. 1989;17:391–7. [PubMed] [Google Scholar]
  • 87.Jelkmann W. Use of recombinant human erythropoietin as an antianemic and performance enhancing drug. Curr Pharm Biotechnol. 2000;1:11–31. doi: 10.2174/1389201003379068. [DOI] [PubMed] [Google Scholar]
  • 88.Sasaki R, Masuda S, Nagao M. Pleiotropic functions and tissue-specific expression of erythropoietin. News Physiol Sci. 2001;16:110–3. doi: 10.1152/physiologyonline.2001.16.3.110. [DOI] [PubMed] [Google Scholar]
  • 89.Sirèn AL, Knerlich F, Poser W, et al. Erythropoietin and erythropoietin receptor in human ischemic/hypoxic brain. Acta Neuropathol. 2001;101:271–6. doi: 10.1007/s004010000297. [DOI] [PubMed] [Google Scholar]
  • 90.Youssoufian H, Longmore G, Neumann D, et al. Structure, function, and activation of the erythropoietin receptor. Blood. 1993;81:2223–36. [PubMed] [Google Scholar]
  • 91.Yu X, Shacka JJ, Eells JB, et al. Erythropoietin receptor signalling is required for normal brain development. Development. 2002;129:505–16. doi: 10.1242/dev.129.2.505. [DOI] [PubMed] [Google Scholar]
  • 92.Ammarguellat F, Gogusev J, Drüeke TB. Direct effect of erythropoietin on rat vascular smooth-muscle cell via a putative erythropoietin receptor. Nephrol Dial Transplant. 1996;11:687–92. doi: 10.1093/oxfordjournals.ndt.a027361. [DOI] [PubMed] [Google Scholar]
  • 93.Ogilvie M, Yu X, Nicolas-Metral V, et al. Erythropoietin stimulates proliferation and interferes with differentiation of myoblasts. J Biol Chem. 2000;275:39754–61. doi: 10.1074/jbc.M004999200. [DOI] [PubMed] [Google Scholar]
  • 94.Anagnostou A, Liu Z, Steiner M, et al. Erythropoietin receptor mRNA expression in human endothelial cells. Proc Natl Acad Sci USA. 1994;91:3974–8. doi: 10.1073/pnas.91.9.3974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Wu H, Lee SH, Gao J, et al. Inactivation of erythropoietin leads to defects in cardiac morphogenesis. Development. 1999;126:3597–605. doi: 10.1242/dev.126.16.3597. [DOI] [PubMed] [Google Scholar]
  • 96.Westenfelder C, Biddle DL, Baranowski RL. Human, rat, and mouse kidney cells express functional erythropoietin receptors. Kidney Int. 1999;55:808–20. doi: 10.1046/j.1523-1755.1999.055003808.x. [DOI] [PubMed] [Google Scholar]
  • 97.Masuda S, Nagao M, Takahata K, et al. Functional erythropoietin receptor of the cells with neural characteristics. Comparison with receptor properties of erythroid cells. J Biol Chem. 1993;268:11208–16. [PubMed] [Google Scholar]
  • 98.Morishita E, Masuda S, Nagao M, et al. Erythropoietin receptor is expressed in rat hippocampal and cerebral cortical neurons and erythropoietin prevents in vitro glutamate-induced neuronal death. Neurosci. 1997;76:105–16. doi: 10.1016/s0306-4522(96)00306-5. [DOI] [PubMed] [Google Scholar]
  • 99.Liu C, Shen K, Liu Z, Noguchi CT. Regulated human erythropoietin receptor expression in mouse brain. J Biol Chem. 1997;272:32395–400. doi: 10.1074/jbc.272.51.32395. [DOI] [PubMed] [Google Scholar]
  • 100.Bouscary D, Pene F, Claessens YE, et al. Critical role for PI 3-kinase in the control of erythropoietin-induced erythroid progenitor proliferation. Blood. 2003;101:3436–43. doi: 10.1182/blood-2002-07-2332. [DOI] [PubMed] [Google Scholar]
  • 101.Wojchowski DM, Gregory RC, Miller CP, et al. Signal transduction in the erythropoietin receptor system. Exp Cell Res. 1999;253:143–56. doi: 10.1006/excr.1999.4673. [DOI] [PubMed] [Google Scholar]
  • 102.Silva M, Grillot D, Benito A, et al. Erythropoietin can promote erythroid progenitor survival by repressing apoptosis through Bcl-XL and Bcl-2. Blood. 1996;88:1576–82. [PubMed] [Google Scholar]
  • 103.Stohlawetz PJ, Dzirlo L, Hergovich N, et al. Effects of erythropoietin on platelet reactivity and thrombopoiesis in humans. Blood. 2000;95:2983–9. [PubMed] [Google Scholar]
  • 104.Zhou XJ, Vaziri ND. Defective calcium signalling in uraemic platelets and its amelioration with long-term erythropoietin therapy. Nephrol Dial Transplant. 2002;17:992–7. doi: 10.1093/ndt/17.6.992. [DOI] [PubMed] [Google Scholar]
  • 105.Bokemeyer C, Aapro MS, Courdi A, et al. European Organisation for Research and Treatment of Cancer (EORTC) Taskforce for the Elderly. EORTC guidelines for the use of erythropoietic proteins in anaemic patients with cancer: 2006 update. Eur J Cancer. 2007;43:258–70. doi: 10.1016/j.ejca.2006.10.014. [DOI] [PubMed] [Google Scholar]
  • 106.Leist M, Ghezzi P, Grasso G, et al. Derivatives of erythropoietin that are tissue protective but not erythropoietic. Science. 2004;305:239–42. doi: 10.1126/science.1098313. [DOI] [PubMed] [Google Scholar]
  • 107.Sirèn AL, Fasshauer T, Bartels C, et al. Therapeutic potential of erythropoietin and its structural or functional variants in the nervous system. Neurotherapeutics. 2009;6:108–27. doi: 10.1016/j.nurt.2008.10.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Dame C, Bartmann P, Wolber E, et al. Erythropoietin gene expression in different areas of the developing human central nervous system. Brain Res Dev Brain Res. 2000;125:69–74. doi: 10.1016/s0165-3806(00)00118-8. [DOI] [PubMed] [Google Scholar]
  • 109.Lewczuk P, Hasselblatt M, Kamrowski-Kruck H, et al. Survival of hippocampal neurons in culture upon hypoxia: effect of erythropoietin. Neuroreport. 2000;11:3485–8. doi: 10.1097/00001756-200011090-00017. [DOI] [PubMed] [Google Scholar]
  • 110.Sirèn AL, Fratelli M, Brines M, et al. Erythropoietin prevents neuronal apoptosis after cerebral ischemia and metabolic stress. Proc Natl Acad Sci USA. 2001;98:4044–9. doi: 10.1073/pnas.051606598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Sinor AD, Greenberg DA. Erythropoietin protects cultured cortical neurons, but not astroglia, from hypoxia and AMPA toxicity. Neurosci Lett. 2000;290:213–5. doi: 10.1016/s0304-3940(00)01361-6. [DOI] [PubMed] [Google Scholar]
  • 112.Dzietko M, Felderhoff-Mueser U, Sifringer M, et al. Erythropoietin protects the developing brain against N-methyl–aspartate receptor antagonist neurotoxicity. Neurobiol Dis. 2004;15:177–87. doi: 10.1016/j.nbd.2003.10.006. [DOI] [PubMed] [Google Scholar]
  • 113.Eid T, Brines ML, Cerami A, et al. Increased expression of erythropoietin receptor on blood vessels in the human epileptogenic hippocampus with sclerosis. J Neuropathol Exp Neurol. 2004;63:73–83. doi: 10.1093/jnen/63.1.73. [DOI] [PubMed] [Google Scholar]
  • 114.Dirnagl U, Simon RP, Hallenbeck JM. Ischemic tolerance and endogenous neuroprotection. Trends Neurosci. 2003;26:248–54. doi: 10.1016/S0166-2236(03)00071-7. [DOI] [PubMed] [Google Scholar]
  • 115.Matsushita H, Johnston MV, Lange MS, Wilson MA. Protective effect of erythropoietin in neonatal hypoxic ischemia in mice. Neuro Report. 2003;14:1757–61. doi: 10.1097/00001756-200309150-00020. [DOI] [PubMed] [Google Scholar]
  • 116.Kumral A, Ozer E, Yilmaz O, et al. Neuroprotective effect of erythropoietin on hypoxic–ischemic brain injury in neonatal rats. Biol Neonate. 2003;83:224–8. doi: 10.1159/000068926. [DOI] [PubMed] [Google Scholar]
  • 117.Kumral A, Genc S, Ozer E, et al. Erythropoietin downregulates bax and DP5 proapoptotic gene expression in neonatal hypoxic-ischemic brain injury. Biol Neonate. 2006;89:205–10. doi: 10.1159/000089951. [DOI] [PubMed] [Google Scholar]
  • 118.van der Kooij MA, Groenendaal F, Kavelaars A, et al. Combination of deferoxamine and erythropoietin: therapy for hypoxia-ischemia-induced brain injury in the neonatal rat. Neurosci Lett. 2009;451:109–13. doi: 10.1016/j.neulet.2008.12.013. [DOI] [PubMed] [Google Scholar]
  • 119.Wen TC, Sadamoto Y, Tanaka J, et al. Erythropoietin protects neurons against chemical hypoxia and cerebral ischemic injury by up-regulating Bcl-xL expression. J Neurosci Res. 2002;67:795–803. doi: 10.1002/jnr.10166. [DOI] [PubMed] [Google Scholar]
  • 120.Villa P, Bigini P, Mennini T, et al. Erythropoietin selectively attenuates cytokine production and inflammation in cerebral ischemia by targeting neuronal apoptosis. J Exp Med. 2003;198:971–5. doi: 10.1084/jem.20021067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Villa P, van Beek J, Larsen AK, et al. Reduced functional deficits, neuroinflammation, and secondary tissue damage after treatment of stroke by nonerythropoietic erythropoietin derivatives. J Cereb Blood Flow Metab. 2007;27:552–63. doi: 10.1038/sj.jcbfm.9600370. [DOI] [PubMed] [Google Scholar]
  • 122.Verdonck O, Lahrech H, Francony G, et al. Erythropoietin protects from post-traumatic edema in the rat brain. J Cereb Blood Flow Metab. 2007;27:1369–76. doi: 10.1038/sj.jcbfm.9600443. [DOI] [PubMed] [Google Scholar]
  • 123.Gunnarson E, Song Y, Kowalewski JM, et al. Erythropoietin modulation of astrocyte water permeability as a component of neuroprotection. Proc Natl Acad Sci USA. 2009;106:1602–7. doi: 10.1073/pnas.0812708106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Chattopadhyay A, Choudhury TD, Bandyopadhyay D, et al. Protective effect of erythropoietin on the oxidative damage of erythrocyte membrane by hydroxyl radical. Biochem Pharmacol. 2000;59:419–25. doi: 10.1016/s0006-2952(99)00277-4. [DOI] [PubMed] [Google Scholar]
  • 125.Genc S, Akhisaroglu M, Kuralay F, et al. Erythropoietin restores glutathione peroxidase activity in 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine-induced neurotoxicity in C57BL mice and stimulates murine astroglial glutathione peroxidase production in vitro. Neurosci Lett. 2002;15:73–6. doi: 10.1016/s0304-3940(02)00041-1. [DOI] [PubMed] [Google Scholar]
  • 126.Agnello D, Bigini P, Villa P, et al. Erythropoietin exerts an anti-inflammatory effect on the CNS in a model of experimental autoimmune encephalomyelitis. Brain Res. 2002;952:128–34. doi: 10.1016/s0006-8993(02)03239-0. [DOI] [PubMed] [Google Scholar]
  • 127.Kawakami M, Sekiguchi M, Sato K, et al. Erythropoietin receptor-mediated inhibition of exocytotic glutamate release confers neuroprotection during chemical ischemia. J Biol Chem. 2001;276:39469–75. doi: 10.1074/jbc.M105832200. [DOI] [PubMed] [Google Scholar]
  • 128.Campana WM, Myers RR. Exogenous erythropoietin protects against dorsal root ganglion apoptosis and pain following peripheral nerve injury. Eur J Neurosci. 2003;18:1497–506. doi: 10.1046/j.1460-9568.2003.02875.x. [DOI] [PubMed] [Google Scholar]
  • 129.Jelkmann W. Effects of erythropoietin on brain function. Curr Pharm Biotechnol. 2005;6:65–79. doi: 10.2174/1389201053167257. [DOI] [PubMed] [Google Scholar]
  • 130.Yamaji R, Okada T, Moriya M, et al. Brain capillary endothelial cells express two forms of erythropoietin receptor mRNA. Eur J Biochem. 1996;239:494–500. doi: 10.1111/j.1432-1033.1996.0494u.x. [DOI] [PubMed] [Google Scholar]
  • 131.Shingo T, Sorokan ST, Shimazaki T, et al. Erythropoietin regulates the in vitro and in vivo production of neuronal progenitors by mammalian forebrain neural stem cells. J Neurosci. 2001;21:9733–43. doi: 10.1523/JNEUROSCI.21-24-09733.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Minnerup J, Heidrich J, Rogalewski A, et al. The efficacy of erythropoietin and its analogues in animal stroke models: a meta-analysis. Stroke. 2009;40:3113–20. doi: 10.1161/STROKEAHA.109.555789. [DOI] [PubMed] [Google Scholar]
  • 133.Brines M, Grasso G, Fiordaliso F, et al. Erythropoietin mediates tissue protection through an erythropoietin and common beta-subunit heteroreceptor. Proc Natl Acad Sci USA. 2004;101:14907–12. doi: 10.1073/pnas.0406491101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Hasselblatt M, Ehrenreich H, Siren AL. The brain erythropoietin system and its potential for therapeutic exploitation in brain disease. J Neurosurg Anesthesiol. 2006;18:132–8. doi: 10.1097/00008506-200604000-00007. [DOI] [PubMed] [Google Scholar]
  • 135.Brines M, Cerami A. Emerging biological roles for erythropoietin in the nervous system. Nat Rev Neurosci. 2005;6:484–94. doi: 10.1038/nrn1687. [DOI] [PubMed] [Google Scholar]
  • 136.Arcasoy MO. The non-haematopoietic biological effects of erythropoietin. Br J Haematol. 2008;141:14–31. doi: 10.1111/j.1365-2141.2008.07014.x. [DOI] [PubMed] [Google Scholar]
  • 137.Kumral A, Baskin H, Yesilirmak DC, et al. Erythropoietin attenuates lipopolysaccharide-induced white matter injury in the neonatal rat brain. Neonatology. 2007;92:269–78. doi: 10.1159/000105493. [DOI] [PubMed] [Google Scholar]
  • 138.Ehrenreich H, Hasselblatt M, Dembowski C, et al. Erythropoietin therapy for acute stroke is both safe and beneficial. Mol Med. 2002;8:495–505. [PMC free article] [PubMed] [Google Scholar]
  • 139.Sirèn AL, Fasshauer T, Bartels C, et al. Therapeutic potential of erythropoietin and its structural or functional variants in the nervous system. Neurotherapeutics. 2009;6:108–27. doi: 10.1016/j.nurt.2008.10.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Huebner K, Isobe M, Croce CM, et al. The human gene encoding GM-CSF is at 5q21-q32, the chromosome region deleted in the 5q- anomaly. Science. 1985;230:1282–5. doi: 10.1126/science.2999978. [DOI] [PubMed] [Google Scholar]
  • 141.Martinez-Moczygemba M, Huston DP. Biology of common beta receptor-signaling cytokines: IL-3, IL-5, and GM-CSF. J Allergy Clin Immunol. 2003;112:653–65. doi: 10.1016/S0091. [DOI] [PubMed] [Google Scholar]
  • 142.Metcalf D. The molecular biology and functions of the granulocyte-macrophage colony-stimulating factors. Blood. 1986;67:257–67. [PubMed] [Google Scholar]
  • 143.Fleetwood AJ, Cook AD, Hamilton JA. Functions of granulocyte-macrophage colony-stimulating factor. Crit Rev Immunol. 2005;25:405–28. doi: 10.1615/critrevimmunol.v25.i5.50. [DOI] [PubMed] [Google Scholar]
  • 144.Conti L, Gessani S. GM-CSF in the generation of dendritic cells from human blood monocyte precursors: recent advances. Immunobiology. 2008;213:859–70. doi: 10.1016/j.imbio.2008.07.017. [DOI] [PubMed] [Google Scholar]
  • 145.de Groot RP, Coffer PJ, Koenderman L. Regulation of proliferation, differentiation and survival by the IL-3/IL-5/GM-CSF receptor family. Cell Signal. 1998;10:619–28. doi: 10.1016/s0898-6568(98)00023-0. [DOI] [PubMed] [Google Scholar]
  • 146.Hercus TR, Thomas D, Guthridge MA, et al. The granulocyte-macrophage colony-stimulating factor receptor: linking its structure to cell signaling and its role in disease. Blood. 2009;114:1289–98. doi: 10.1182/blood-2008-12-164004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Schuster SJ, Venugopal P, Kern JC, et al. GM-CSF plus rituximab immunotherapy: translation of biologic mechanisms into therapy for indolent B-cell lymphomas. Leuk Lymphoma. 2008;49:1681–92. doi: 10.1080/10428190802216731. [DOI] [PubMed] [Google Scholar]
  • 148.Wadhwa M, Thorpe R. Haematopoietic growth factors and their therapeutic use. Thromb Haemost. 2008;99:863–73. [PubMed] [Google Scholar]
  • 149.Schuster SJ, Venugopal P, Kern JC, et al. GM-CSF plus rituximab immunotherapy: translation of biologic mechanisms into therapy for indolent B-cell lymphomas. Leuk Lymphoma. 2008;49:1681–92. doi: 10.1080/10428190802216731. [DOI] [PubMed] [Google Scholar]
  • 150.Elias EG, Zapas JL, McCarron EC, et al. Sequential administration of GM-CSF (Sargramostim) and IL-2 +/− autologous vaccine as adjuvant therapy in cutaneous melanoma: an interim report of a phase II clinical trial. Cancer Biother Radiopharm. 2008;23:285–91. doi: 10.1089/cbr.2007.0438. [DOI] [PubMed] [Google Scholar]
  • 151.Safdar A, Rodriguez GH, Rueda AM, et al. Multiple-dose granulocyte-macrophage-colony-stimulating factor plus 23-valent polysaccharide pneumococcal vaccine in patients with chronic lymphocytic leukemia: a prospective, randomized trial of safety and immunogenicity. Cancer. 2008;113:383–7. doi: 10.1002/cncr.23561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Lieschke GJ, Maher D, Cebon J, et al. Effects of bacterially synthesized recombinant human granulocyte-macrophage colony-stimulating factor in patients with advanced malignancy. Ann Intern Med. 1989;110:357–64. doi: 10.7326/0003-4819-110-5-357. [DOI] [PubMed] [Google Scholar]
  • 153.Tarkowski E, Rosengren L, Blomstrand C, et al. Intrathecal release of pro- and anti-inflammatory cytokines during stroke. Clin Exp Immunol. 1997;110:492–9. doi: 10.1046/j.1365-2249.1997.4621483.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Tarkowski E, Rosengren L, Blomstrand C, et al. Intrathecal expression of proteins regulating apoptosis in acute stroke. Stroke. 1999;30:321–7. doi: 10.1161/01.str.30.2.321. [DOI] [PubMed] [Google Scholar]
  • 155.Schäbitz WR, Krüger C, Pitzer C, et al. A neuroprotective function for the hematopoietic protein granulocyte-macrophage colony stimulating factor (GM-CSF) J Cereb Blood Flow Metab. 2008;28:29–43. doi: 10.1038/sj.jcbfm.9600496. [DOI] [PubMed] [Google Scholar]
  • 156.Krüger C, Laage R, Pitzer C, et al. The hematopoietic factor GM-CSF (granulocyte-macrophage colony-stimulating factor) promotes neuronal differentiation of adult neural stem cells in vitro. BMC Neurosci. 2007;8:88. doi: 10.1186/1471-2202-8-88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Nakagawa T, Suga S, Kawase T, et al. Intracarotid injection of granulocyte-macrophage colony-stimulating factor induces neuroprotection in a rat transient middle cerebral artery occlusion model. Brain Res. 2006;1089:179–85. doi: 10.1016/j.brainres.2006.03.059. [DOI] [PubMed] [Google Scholar]
  • 158.Todo K, Kitagawa K, Sasaki T, et al. Granulocyte-macrophage colony-stimulating factor enhances leptomeningeal collateral growth induced by common carotid artery occlusion. Stroke. 2008;39:1875–82. doi: 10.1161/STROKEAHA.107.503433. [DOI] [PubMed] [Google Scholar]
  • 159.Kong T, Choi JK, Park H, et al. Reduction in programmed cell death and improvement in functional outcome of transient focal cerebral ischemia after administration of granulocyte-macrophage colony-stimulating factor in rats. J Neurosurg. 2009;111:155–63. doi: 10.3171/2008.12.JNS08172. [DOI] [PubMed] [Google Scholar]
  • 160.Yata K, Matchett GA, Tsubokawa T, et al. Granulocyte-colony stimulating factor inhibits apoptotic neuron loss after neonatal hypoxia-ischemia in rats. Brain Res. 2007;1145:227–38. doi: 10.1016/j.brainres.2007.01.144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Buschmann IR, Busch HJ, Mies G, et al. Therapeutic induction of arteriogenesis in hypoperfused rat brain via granulocyte-macrophage colony-stimulating factor. Circulation. 2003;108:610–5. doi: 10.1161/01.CIR.0000074209.17561.99. [DOI] [PubMed] [Google Scholar]
  • 162.Schneeloch E, Mies G, Busch HJ, et al. Granulocyte–macrophage colony-stimulating factor-induced arteriogenesis reduces energy failure in hemodynamic stroke. Proc Natl Acad Sci USA. 2004;101:12730–5. doi: 10.1073/pnas.0404880101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Schneider UC, Schilling L, Schroeck H, et al. Granulocyte-macrophage colony-stimulating factor-induced vessel growth restores cerebral blood supply after bilateral carotid artery occlusion. Stroke. 2007;38:1320–8. doi: 10.1161/01.STR.0000259707.43496.71. [DOI] [PubMed] [Google Scholar]
  • 164.Love R. GM-CSF induced arteriogenesis: a potential treatment for stroke? Lancet Neurol. 2003;2:458. doi: 10.1016/s1474-4422(03)00478-2. [DOI] [PubMed] [Google Scholar]
  • 165.Paczkowska E, Larysz B, Rzeuski R, et al. Human hematopoietic stem/progenitor-enriched CD34(+) cells are mobilized into peripheral blood during stress related to ischemic stroke or acute myocardial infarction. Eur J Haematol. 2005;75:461–7. doi: 10.1111/j.1600-0609.2005.00536.x. [DOI] [PubMed] [Google Scholar]
  • 166.Navarro M, Rosell A, Penalba A, et al. Role of endogenous granulocyte-macrophage colony stimulating factor following stroke and relationship to neurological outcome. Curr Neurovasc Res. 2009;6:246–51. doi: 10.2174/156720209789630366. [DOI] [PubMed] [Google Scholar]
  • 167.Breen EC. VEGF in biological control. J Cell Biochem. 2007;102:1358–67. doi: 10.1002/jcb.21579. [DOI] [PubMed] [Google Scholar]
  • 168.Leung DW, Cachianes G, Kuang WJ, et al. Vascular endothelial growth factor is a secreted angiogenic mitogen. Science. 1989;246:1306–9. doi: 10.1126/science.2479986. [DOI] [PubMed] [Google Scholar]
  • 169.Monacci WT, Merrill MJ, Oldfield EH. Expression of vascular permeability factor/vascular endothelial growth factor in normal rat tissues. Am J Physiol. 1993;264:C995–1002. doi: 10.1152/ajpcell.1993.264.4.C995. [DOI] [PubMed] [Google Scholar]
  • 170.Marti HH, Risau W. Angiogenesis in ischemic disease. Thromb Haemost. 1999;82:44–52. [PubMed] [Google Scholar]
  • 171.Costa C, Soares R, Schmitt F. Angiogenesis: now and then. APMIS. 2004;112:402–12. doi: 10.1111/j.1600-0463.2004.apm11207-0802.x. [DOI] [PubMed] [Google Scholar]
  • 172.Ellis LM, Staley CA, Liu W, et al. Down-regulation of vascular endothelial growth factor in a human colon carcinoma cell line transfected with an antisense expression vector specific for c-src. J Biol Chem. 1998;273:1052–7. doi: 10.1074/jbc.273.2.1052. [DOI] [PubMed] [Google Scholar]
  • 173.Ebos JM, Tran J, Master Z, et al. Imatinib mesylate (STI-571) reduces Bcr-Abl-mediated vascular endothelial growth factor secretion in chronic myelogenous leukemia. Mol Cancer Res. 2002;1:89–95. [PubMed] [Google Scholar]
  • 174.Ikeda N, Nakajima Y, Sho M, et al. The association of K-ras gene mutation and vascular endothelial growth factor gene expression in pancreatic carcinoma. Cancer. 2001;92:488–99. doi: 10.1002/1097-0142(20010801)92:3<488::aid-cncr1347>3.0.co;2-f. [DOI] [PubMed] [Google Scholar]
  • 175.Fujisawa T, Watanabe J, Kamata Y, et al. Effect of p53 gene transfection on vascular endothelial growth factor expression in endometrial cancer cells. Exp Mol Pathol. 2003;74:276–81. doi: 10.1016/s0014-4800(03)00020-0. [DOI] [PubMed] [Google Scholar]
  • 176.Joo YE, Rew JS, Seo YH, et al. Cyclooxygenase-2 overexpression correlates with vascular endothelial growth factor expression and tumor angiogenesis in gastric cancer. J Clin Gastroenterol. 2003;37:28–33. doi: 10.1097/00004836-200307000-00009. [DOI] [PubMed] [Google Scholar]
  • 177.Dong J, Grunstein J, Tejada M, et al. VEGF-null cells require PDGFR alpha signaling-mediated stromal fibroblast recruitment for tumorigenesis. EMBO J. 2004;23:2800–10. doi: 10.1038/sj.emboj.7600289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Olsson AK, Dimberg A, Kreuger J, et al. VEGF receptor signalling – in control of vascular function. Nat Rev Mol Cell Biol. 2006;7:359–71. doi: 10.1038/nrm1911. [DOI] [PubMed] [Google Scholar]
  • 179.Autiero M, Waltenberger J, Communi D, et al. Role of PlGF in the intra- and intermolecular cross talk between the VEGF receptors Flt1 and Flk1. Nat Med. 2003;9:936–43. doi: 10.1038/nm884. [DOI] [PubMed] [Google Scholar]
  • 180.Foster RR, Hole R, Anderson K, et al. Functional evidence that vascular endothelial growth factor may act as an autocrine factor on human podocytes. Am J Physiol Renal Physiol. 2003;284:F1263–73. doi: 10.1152/ajprenal.00276.2002. [DOI] [PubMed] [Google Scholar]
  • 181.Oosthuyse B, Moons L, Storkebaum E, et al. Deletion of the hypoxia-response element in the vascular endothelial growth factor promoter causes motor neuron degeneration. Nat Genet. 2001;28:131–8. doi: 10.1038/88842. [DOI] [PubMed] [Google Scholar]
  • 182.Svensson B, Peters M, König HG, et al. Vascular endothelial growth factor protects cultured rat hippocampal neurons against hypoxic injury via an antiexcitotoxic, caspase-independent mechanism. J Cereb Blood Flow Metab. 2002;22:1170–5. doi: 10.1097/01.wcb.0000037988.07114.98. [DOI] [PubMed] [Google Scholar]
  • 183.Schänzer A, Wachs FP, Wilhelm D, et al. Direct stimulation of adult neural stem cells in vitro and neurogenesis in vivo by vascular endothelial growth factor. Brain Pathol. 2004;14:237–48. doi: 10.1111/j.1750-3639.2004.tb00060.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Jin K, Mao XO, Eshoo MW, et al. Microarray analysis of hippocampal gene expression in global cerebral ischemia. Ann Neurol. 2001;50:93–103. doi: 10.1002/ana.1073. [DOI] [PubMed] [Google Scholar]
  • 185.Marti HJ, Bernaudin M, Bellail A, et al. Hypoxia-induced vascular endothelial growth factor expression precedes neovascularization after cerebral ischemia. Am J Pathol. 2000;156:965–76. doi: 10.1016/S0002-9440(10)64964-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Kovacs Z, Ikezaki K, Samoto K, et al. VEGF and flt expression time kinetics in rat brain infarct. Stroke. 1996;27:1865–73. doi: 10.1161/01.str.27.10.1865. [DOI] [PubMed] [Google Scholar]
  • 187.Hayashi T, Abe K, Suzuki H, et al. Rapid induction of vascular endothelial growth factor gene expression after transient middle cerebral artery occlusion in rats. Stroke. 1997;28:2039–44. doi: 10.1161/01.str.28.10.2039. [DOI] [PubMed] [Google Scholar]
  • 188.Hayashi T, Noshita N, Sugawara T, et al. Temporal profile of angiogenesis and expression of related genes in the brain after ischemia. J Cereb Blood Flow Metab. 2003;23:166–80. doi: 10.1097/01.WCB.0000041283.53351.CB. [DOI] [PubMed] [Google Scholar]
  • 189.Forsythe JA, Jiang BH, Iyer NV, et al. Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1. Mol Cell Biol. 1996;16:4604–13. doi: 10.1128/mcb.16.9.4604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Ema M, Taya S, Yokotani N, et al. A novel bHLH-PAS factor with close sequence similarity to hypoxia-inducible factor 1alpha regulates the VEGF expression and is potentially involved in lung and vascular development. Proc Natl Acad Sci USA. 1997;94:4273–8. doi: 10.1073/pnas.94.9.4273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Weis SM, Cheresh DA. Pathophysiological consequences of VEGF-induced vascular permeability. Nature. 2005;437:497–504. doi: 10.1038/nature03987. [DOI] [PubMed] [Google Scholar]
  • 192.Tang Y, Pacary E, Frèret T, et al. Effect of hypoxic preconditioning on brain genomic response before and following ischemia in the adult mouse: identification of potential neuroprotective candidates for stroke. Neurobiol Dis. 2006;21:18–28. doi: 10.1016/j.nbd.2005.06.002. [DOI] [PubMed] [Google Scholar]
  • 193.Gu W, Brännström T, Jiang W, et al. Vascular endothelial growth factor-A and -C protein up-regulation and early angiogenesis in a rat photothrombotic ring stroke model with spontaneous reperfusion. Acta Neuropathol. 2001;102:216–26. doi: 10.1007/s004010100370. [DOI] [PubMed] [Google Scholar]
  • 194.Lafuente JV, Bulnes S, Mitre B, et al. Role of VEGF in an experimental model of cortical micronecrosis. Amino Acids. 2002;23:241–5. doi: 10.1007/s00726-001-0135-1. [DOI] [PubMed] [Google Scholar]
  • 195.Leker RR, Teichner A, Ovadia H, et al. Expression of endothelial nitric oxide synthase in the ischemic penumbra: relationship to expression of neuronal nitric oxide synthase and vascular endothelial growth factor. Brain Res. 2001;909:1–7. doi: 10.1016/s0006-8993(01)02561-6. [DOI] [PubMed] [Google Scholar]
  • 196.Zhang R, Wang L, Zhang L, et al. Nitric oxide enhances angiogenesis via the synthesis of vascular endothelial growth factor and cGMP after stroke in the rat. Circ Res. 2003;92:308–13. doi: 10.1161/01.res.0000056757.93432.8c. [DOI] [PubMed] [Google Scholar]
  • 197.Wang Y, Kilic E, Kilic U, et al. VEGF overexpression induces post-ischaemic neuroprotection, but facilitates haemodynamic steal phenomena. Brain. 2005;128:52–63. doi: 10.1093/brain/awh325. [DOI] [PubMed] [Google Scholar]
  • 198.Wang Y, Jin K, Mao XO, et al. VEGF-overexpressing transgenic mice show enhanced post-ischemic neurogenesis and neuromigration. J Neurosci Res. 2007;85:740–7. doi: 10.1002/jnr.21169. [DOI] [PubMed] [Google Scholar]
  • 199.Harrigan MR, Ennis SR, Sullivan SE, et al. Effects of intraventricular infusion of vascular endothelial growth factor on cerebral blood flow, edema, and infarct volume. Acta Neurochir. 2003;145:49–53. doi: 10.1007/s00701-002-1035-1. [DOI] [PubMed] [Google Scholar]
  • 200.Hayashi T, Abe K, Itoyama Y. Reduction of ischemic damage by application of vascular endothelial growth factor in rat brain after transient ischemia. J Cereb Blood Flow Metab. 1998;18:887–95. doi: 10.1097/00004647-199808000-00009. [DOI] [PubMed] [Google Scholar]
  • 201.Foster KA, Regan HK, Danziger AP, et al. Attenuation of edema and infarct volume following focal cerebral ischemia by early but not delayed administration of a novel small molecule KDR kinase inhibitor. Neurosci Res. 2009;63:10–6. doi: 10.1016/j.neures.2008.09.007. [DOI] [PubMed] [Google Scholar]
  • 202.Kaya D, Gürsoy-Ozdemir Y, Yemisci M, et al. VEGF protects brain against focal ischemia without increasing blood – brain permeability when administered intracerebroventricularly. J Cereb Blood Flow Metab. 2005;25:1111–8. doi: 10.1038/sj.jcbfm.9600109. [DOI] [PubMed] [Google Scholar]
  • 203.Zhang ZG, Zhang L, Jiang Q, et al. VEGF enhances angiogenesis and promotes blood-brain barrier leakage in the ischemic brain. J Clin Invest. 2000;106:829–38. doi: 10.1172/JCI9369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Wang Y, Galvan V, Gorostiza O, et al. Vascular endothelial growth factor improves recovery of sensorimotor and cognitive deficits after focal cerebral ischemia in the rat. Brain Res. 2006;1115:186–93. doi: 10.1016/j.brainres.2006.07.060. [DOI] [PubMed] [Google Scholar]
  • 205.Chu K, Park KI, Lee ST, et al. Combined treatment of vascular endothelial growth factor and human neural stem cells in experimental focal cerebral ischemia. Neurosci Res. 2005;53:384–90. doi: 10.1016/j.neures.2005.08.010. [DOI] [PubMed] [Google Scholar]
  • 206.Beck H, Acker T, Wiessner C, et al. Expression of angiopoietin-1, angiopoietin-2, and tie receptors after middle cerebral artery occlusion in the rat. Am J Pathol. 2000;157:1473–83. doi: 10.1016/S0002-9440(10)64786-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Zhu Y, Lee C, Shen F, et al. Angiopoietin-2 facilitates vascular endothelial growth factor-induced angiogenesis in the mature mouse brain. Stroke. 2005;36:1533–7. doi: 10.1161/01.STR.0000170712.46106.2e. [DOI] [PubMed] [Google Scholar]
  • 208.Zhang ZG, Zhang L, Tsang W, et al. Correlation of VEGF and angiopoietin expression with disruption of blood-brain barrier and angiogenesis after focal cerebral ischemia. J Cereb Blood Flow Metab. 2002;22:379–92. doi: 10.1097/00004647-200204000-00002. [DOI] [PubMed] [Google Scholar]
  • 209.Zhang Z, Chopp M. Vascular endothelial growth factor and angiopoietins in focal cerebral ischemia. Trends Cardiovasc Med. 2002;12:62–6. doi: 10.1016/s1050-1738(01)00149-9. [DOI] [PubMed] [Google Scholar]
  • 210.Zacharek A, Chen J, Cui X, et al. Angiopoietin1/Tie2 and VEGF/Flk1 induced by MSC treatment amplifies angiogenesis and vascular stabilization after stroke. J Cereb Blood Flow Metab. 2007;27:1684–91. doi: 10.1038/sj.jcbfm.9600475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Schoch HJ, Fischer S, Marti HH. Hypoxia-induced vascular endothelial growth factor expression causes vascular leakage in the brain. Brain. 2002;125:2549–57. doi: 10.1093/brain/awf257. [DOI] [PubMed] [Google Scholar]
  • 212.Kimura R, Nakase H, Tamaki R, et al. Vascular endothelial growth factor antagonist reduces brain edema formation and venous infarction. Stroke. 2005;36:1259–63. doi: 10.1161/01.STR.0000165925.20413.14. [DOI] [PubMed] [Google Scholar]
  • 213.Rite I, Machado A, Cano J, et al. Intracerebral VEGF injection highly upregulates AQP4 mRNA and protein in the perivascular space and glia limitans externa. Neurochem Int. 2008;52:897–903. doi: 10.1016/j.neuint.2007.10.004. [DOI] [PubMed] [Google Scholar]
  • 214.Bellomo M, Adamo EB, Deodato B, et al. Enhancement of expression of vascular endothelial growth factor after adeno-associated virus gene transfer is associated with improvement of brain ischemia injury in the gerbil. Pharmacol Res. 2003;48:309–17. doi: 10.1016/s1043-6618(03)00128-2. [DOI] [PubMed] [Google Scholar]
  • 215.Li Z, Wang R, Li S, et al. Intraventricular pre-treatment with rAAV-VEGF induces intracranial hypertension and aggravates ischemic injury at the early stage of transient focal cerebral ischemia in rats. Neurol Res. 2008;30:868–75. doi: 10.1179/174313208X309720. [DOI] [PubMed] [Google Scholar]
  • 216.Manoonkitiwongsa PS, Schultz RL, Whitter EF, et al. Contraindications of VEGF-based therapeutic angiogenesis: effects on macrophage density and histology of normal and ischemic brains. Vascul Pharmacol. 2006;44:316–25. doi: 10.1016/j.vph.2006.01.008. [DOI] [PubMed] [Google Scholar]
  • 217.Stowe AM, Plautz EJ, Nguyen P, et al. Neuronal HIF-1 alpha protein and VEGFR-2 immunoreactivity in functionally related motor areas following a focal M1 infarct. J Cereb Blood Flow Metab. 2008;28:612–20. doi: 10.1038/sj.jcbfm.9600560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Clayton JA, Chalothorn D, Faber JE. Vascular endothelial growth factor-a specifies formation of native collaterals and regulates collateral growth in ischemia. Circ Res. 2008;103:1027–36. doi: 10.1161/CIRCRESAHA.108.181115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Maurer MH, Thomas C, Bürgers HF, et al. Transplantation of adult neural progenitor cells transfected with vascular endothelial growth factor rescues grafted cells in the rat brain. Int J Biol Sci. 2007;4:1–7. doi: 10.7150/ijbs.4.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Miki Y, Nonoguchi N, Ikeda N, et al. Vascular endothelial growth factor gene-transferred bone marrow stromal cells engineered with a herpes simplex virus type 1 vector can improve neurological deficits and reduce infarction volume in rat brain ischemia. Neurosurgery. 2007;61:586–94. doi: 10.1227/01.NEU.0000290907.30814.42. [DOI] [PubMed] [Google Scholar]
  • 221.Bleul CC, Farzan M, Choe H, et al. The lymphocyte chemoattractant SDF-1 is a ligand for LESTR/fusin and blocks HIV-1 entry. Nature. 1996b;382:829–33. doi: 10.1038/382829a0. [DOI] [PubMed] [Google Scholar]
  • 222.Feng Y, Broder CC, Kennedy PE, et al. HIV-1 entry cofactor: functional cDNA cloning of a seven-transmembrane, G protein coupled receptor. Science. 1996;272:872–7. doi: 10.1126/science.272.5263.872. [DOI] [PubMed] [Google Scholar]
  • 223.Oberlin E, Amara A, Bachelerie F, et al. The CXC chemokine SDF-1 is the ligand for LESTR/fusin and prevents infection by T-cellline- adapted HIV-1. Nature. 1996;382:833–5. doi: 10.1038/382833a0. [DOI] [PubMed] [Google Scholar]
  • 224.Petit I, Szyper-Kravitz M, Nagler A, et al. G-CSF induces stem cell mobilization by decreasing bone marrow SDF-1 and up-regulating CXCR4. Nat Immunol. 2002;3:687–94. doi: 10.1038/ni813. [DOI] [PubMed] [Google Scholar]
  • 225.Shirozu M, Nakano T, Inazawa J, et al. Structure and chromosomal localization of the human stromal cell-derived factor 1 (SDF1) gene. Genomics. 1995;28:495–500. doi: 10.1006/geno.1995.1180. [DOI] [PubMed] [Google Scholar]
  • 226.Klein RS, Rubin JB, Gibson HD, et al. SDF-1 alpha induces chemotaxis and enhances Sonic hedgehog induced proliferation of cerebellar granule cells. Development. 2001;128:1971–81. doi: 10.1242/dev.128.11.1971. [DOI] [PubMed] [Google Scholar]
  • 227.Stumm RK, Zhou C, Ara T, et al. CXCR4 regulates interneuron migration in the developing neocortex. J Neurosci. 2003;23:5123–30. doi: 10.1523/JNEUROSCI.23-12-05123.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Borrell V, Marin O. Meninges control tangential migration of hem-derived Cajal–Retzius cells via CXCL12/CXCR4 signaling. Nat. Neurosci. 2006;9:1284–93. doi: 10.1038/nn1764. [DOI] [PubMed] [Google Scholar]
  • 229.Lu M, Grove EA, Miller RJ. Abnormal development of the hippocampal dentate gyrus in mice lacking the CXCR4 chemokine receptor. Proc Natl Acad Sci USA. 2002;99:7090–5. doi: 10.1073/pnas.092013799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Lieberam I, Agalliu D, Nagasawa T, et al. A Cxcl12–CXCR4 chemokine signaling pathway defines the initial trajectory of mammalian motor axons. Neuron. 2005;47:667–79. doi: 10.1016/j.neuron.2005.08.011. [DOI] [PubMed] [Google Scholar]
  • 231.Bagri A, Gurney T, He X, et al. The chemokine SDF1 regulates migration of dentate granule cells. Development. 2002;129:4249–60. doi: 10.1242/dev.129.18.4249. [DOI] [PubMed] [Google Scholar]
  • 232.Yamaguchi J, Kusano KF, Masuo O, et al. Stromal cell-derived factor-1 effects on ex vivo expanded endothelial progenitor cell recruitment for ischemic neovascularization. Circulation. 2003;11(107):1322–8. doi: 10.1161/01.cir.0000055313.77510.22. [DOI] [PubMed] [Google Scholar]
  • 233.Ma Q, Jones D, Borghesani PR, et al. Impaired B-lymphopoiesis, myelopoiesis, and derailed cerebellar neuron migration in CXCR4- and SDF-1-deficient mice. Proc Natl Acad Sci USA. 1998;95:9448–53. doi: 10.1073/pnas.95.16.9448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Zou YR, Kottmann AH, Kuroda M, et al. Function of the chemokine receptor CXCR4 in haematopoiesis and in cerebellar development. Nature. 1998;11(393):595–9. doi: 10.1038/31269. [DOI] [PubMed] [Google Scholar]
  • 235.Suzuki Y, Rahman M, Mitsuya H. Diverse transcriptional response of CD4(+) T cells to stromal cell-derived factor (SDF)-1: cell survival promotion and priming effects of SDF-1 on CD4(+) T cells. J Immunol. 2001;167:3064–73. doi: 10.4049/jimmunol.167.6.3064. [DOI] [PubMed] [Google Scholar]
  • 236.Langford D, Sanders VJ, Mallory M, et al. Expression of stromal cell-derived factor 1alpha protein in HIV encephalitis. J Neuroimmunol. 2002;127:115–26. doi: 10.1016/s0165-5728(02)00068-1. [DOI] [PubMed] [Google Scholar]
  • 237.Neuhaus T, Stier S, Totzke G, et al. Stromal cell-derived factor 1alpha (SDF-1alpha) induces gene-expression of early growth response-1 (Egr-1) and VEGF in human arterial endothelial cells and enhances VEGF induced cell proliferation. Cell Prolif. 2003;36:75–86. doi: 10.1046/j.1365-2184.2003.00262.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Banisadr G, Fontanges P, Haour F, et al. Neuroanatomical distribution of CXCR4 in adult rat brain and its localization in cholinergic and dopaminergic neurons. Eur J Neurosci. 2002;16:1661–71. doi: 10.1046/j.1460-9568.2002.02237.x. [DOI] [PubMed] [Google Scholar]
  • 239.Banisadr G, Skrzydelski D, Kitabgi P, et al. Highly regionalized distribution of stromal cell-derived factor-1/CXCL12 in adult rat brain: constitutive expression in cholinergic, dopaminergic and vasopressinergic neurons. Eur J Neurosci. 2003;18:1593–606. doi: 10.1046/j.1460-9568.2003.02893.x. [DOI] [PubMed] [Google Scholar]
  • 240.Callewaere C, Banisadr G, Desarmenien MG, et al. The chemokine SDF-1/CXCL12 modulates the firing pattern of vasopressin neurons and counteracts induced vasopressin. Proc Natl Acad Sci USA. 2006;103:8221–6. doi: 10.1073/pnas.0602620103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Bezzi P, Domercq M, Brambilla L, et al. Volterra A. CXCR4-activated astrocyte glutamateelease via TNFb: amplification by microglia triggers neurotoxicity. Nat Neurosci. 2001;4:702–10. doi: 10.1038/89490. [DOI] [PubMed] [Google Scholar]
  • 242.Shen LH, Li Y, Chen J, et al. Therapeutic benefit of bone marrow stromal cells administered 1 month after stroke. J. Cereb. Blood Flow Metab. 2007;27:6–13. doi: 10.1038/sj.jcbfm.9600311. [DOI] [PubMed] [Google Scholar]
  • 243.Askari AT, Unzek S, Popovic ZB, et al. Effect of stromal-cell-derived factor 1 on stem-cell homing and tissue regeneration in ischaemic cardiomyopathy. Lancet. 2003;362:697–703. doi: 10.1016/S0140-6736(03)14232-8. [DOI] [PubMed] [Google Scholar]
  • 244.Tögel F, Isaac J, Hu Z, et al. Renal SDF-1 signals mobilization and homing of CXCR4-positive cells to the kidney after ischemic injury. Kidney Int. 2005;67:1772–84. doi: 10.1111/j.1523-1755.2005.00275.x. [DOI] [PubMed] [Google Scholar]
  • 245.Liu Z, Geng L, Li R, et al. Frequency modulation of synchronized Ca2+ spikes in cultured hippocampal networks through G-protein-coupled receptors. J Neurosci. 2003;23:4156–63. doi: 10.1523/JNEUROSCI.23-10-04156.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Stumm RK, Rummel J, Junker V, et al. A dual role for the SDF-1/CXCR4 chemokine receptor system in adult brain: isoform-selective regulation of SDF-1 expression modulates CXCR4-dependent neuronal plasticity and cerebral leukocyte recruitment after focal ischemia. J Neurosci. 2002;22:5865–78. doi: 10.1523/JNEUROSCI.22-14-05865.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Hill WD, Hess DC, Martin-Studdard A, et al. SDF-1 (CXCL12) is upregulated in the ischemic penumbra following stroke: association with bone marrow cell homing to injury. JNEN. 2004;63:84–96. doi: 10.1093/jnen/63.1.84. [DOI] [PubMed] [Google Scholar]
  • 248.Imitola J, Park KI, Teng YD, et al. Stem cells: cross-talk and developmental programs. Philos Trans R Soc Lond B Biol Sci. 2004;359:823–37. doi: 10.1098/rstb.2004.1474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Ohab JJ, Fleming S, Blesch A, et al. A neurovascular niche for neurogenesis after stroke. J Neurosci. 2006;26:13007–16. doi: 10.1523/JNEUROSCI.4323-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.Robin AM, Zhang ZG, Wang L, et al. Stromal cell-derived factor 1alpha mediates neural progenitor cell motility after focal cerebral ischemia. J Cereb Blood Flow Metab. 2006;26:125–34. doi: 10.1038/sj.jcbfm.9600172. [DOI] [PubMed] [Google Scholar]
  • 251.Thored P, Arvidsson A, Cacci E, et al. Persistent production of neurons from adult brain stem cells during recovery after stroke. Stem Cells. 2006;24:739–47. doi: 10.1634/stemcells.2005-0281. [DOI] [PubMed] [Google Scholar]
  • 252.Chang YC, Shyu WC, Lin SZ, et al. Regenerative therapy for stroke. Cell Transplant. 2007;16:171–81. [PubMed] [Google Scholar]
  • 253.Wang Y, Deng Y, Zhou GQ. SDF-1alpha/CXCR4-mediated migration of systemically transplanted bone marrow stromal cells towards ischemic brain lesion in a rat model. Brain Res. 2008;1195:104–12. doi: 10.1016/j.brainres.2007.11.068. [DOI] [PubMed] [Google Scholar]
  • 254.Kucia M, Zhang YP, Reca R, et al. Cells enriched in markers of neural tissue-committed stem cells reside in the bone marrow and are mobilized into the peripheral blood following stroke. Leukemia. 2006;20:18–28. doi: 10.1038/sj.leu.2404011. [DOI] [PubMed] [Google Scholar]
  • 255.Shyu WC, Lin SZ, Yen PS, et al. Stromal cell-derived factor-1 promotes neuroprotection, angiogenesis, and mobilization/homing of bone marrow-derived cells in stroke rats. JPET. 2008a;324:834–49. doi: 10.1124/jpet.107.127746. [DOI] [PubMed] [Google Scholar]
  • 256.Shyu WC, Liu DD, Lin SZ, et al. Implantation of olfactory ensheathing cells promotes neuroplasticity in murine models of stroke. J Clin Invest. 2008b;118:2482–95. doi: 10.1172/JCI34363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Schönemeier B, Schulz S, Hoellt V, et al. Enhanced expression of the CXCl12/SDF-1 chemokine receptor CXCR7 after cerebral ischemia in the rat brain. J Neuroimmunol. 2008;198:39–45. doi: 10.1016/j.jneuroim.2008.04.010. [DOI] [PubMed] [Google Scholar]
  • 258.Hassan HT, Zander A. Stem cell factor as a survival and growth factor in human normal and malignant hematopoiesis. Acta Haematol. 1996;95:257–62. doi: 10.1159/000203893. [DOI] [PubMed] [Google Scholar]
  • 259.Corti S, Locatelli F, Strazzer S, et al. Modulated generation of neuronal cells from bone marrow by expansion and mobilization of circulating stem cells with in vivo cytokine treatment. Exp Neurol. 2002;177:443–52. doi: 10.1006/exnr.2002.8004. [DOI] [PubMed] [Google Scholar]
  • 260.Zhao LR, Singhal S, Duan WM, et al. Brain repair by hematopoietic growth factors in a rat model of stroke. Stroke. 2007a;38:2584–91. doi: 10.1161/STROKEAHA.106.476457. [DOI] [PubMed] [Google Scholar]
  • 261.Zhao LR, Berra HH, Duan WM, et al. Beneficial effects of hematopoietic growth factor therapy in chronic ischemic stroke in rats. Stroke. 2007b;38:2804–11. doi: 10.1161/STROKEAHA.107.486217. [DOI] [PubMed] [Google Scholar]
  • 262.Mattson MP. Glutamate and neurotrophic factors in neuronal plasticity and disease. Ann N Y Acad Sci. 2008;1144:97–112. doi: 10.1196/annals.1418.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263.Dicou E. Neurotrophins and neuronal migration in the developing rodent brain. Brain Res Rev. 2009;60:408–17. doi: 10.1016/j.brainresrev.2009.03.001. [DOI] [PubMed] [Google Scholar]
  • 264.Hamano K, Li TS, Kobayashi T, et al. Angiogenesis induced by the implantation of self-bone marrow cells: a new material for therapeutic angiogenesis. Cell Transplant. 2000;9:439–43. doi: 10.1177/096368970000900315. [DOI] [PubMed] [Google Scholar]
  • 265.Chen X, Li Y, Wang L, et al. Ischemic rat brain extracts induce human marrow stromal cell growth factor production. Neuropathology. 2002;22:275–9. doi: 10.1046/j.1440-1789.2002.00450.x. [DOI] [PubMed] [Google Scholar]
  • 266.Kurozumi K, Nakamura K, Tamiya T, et al. Mesenchymal stem cells that produce neurotrophic factors reduce ischemic damage in the rat middle cerebral artery occlusion model. Mol Ther. 2005;11:96–104. doi: 10.1016/j.ymthe.2004.09.020. [DOI] [PubMed] [Google Scholar]
  • 267.Nomura T, Honmou O, Harada K, et al. I.V. infusion of brain-derived neurotrophic factor gene-modified human mesenchymal stem cells protects against injury in a cerebral ischemia model in adult rat. Neuroscience. 2005;136:161–9. doi: 10.1016/j.neuroscience.2005.06.062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268.Dempsey RJ, Kalluri HS. Ischemia-induced neurogenesis: role of growth factors. Neurosurg Clin N Am. 2007;18:183–90. doi: 10.1016/j.nec.2006.10.011. [DOI] [PubMed] [Google Scholar]
  • 269.McAllister AK. Spatially restricted actions of BDNF. Neuron. 2002;36:549–50. doi: 10.1016/s0896-6273(02)01063-2. [DOI] [PubMed] [Google Scholar]
  • 270.Schäbitz WR, Sommer C, Zoder W, et al. Intravenous brain-derived neurotrophic factor reduces infarct size and counterregulates Bax and Bcl-2 expression after temporary focal cerebral ischemia. Stroke. 2000;31:2212–7. doi: 10.1161/01.str.31.9.2212. [DOI] [PubMed] [Google Scholar]
  • 271.Schäbitz WR, Berger C, Kollmar R, et al. Effect of brain-derived neurotrophic factor treatment and forced arm use on functional motor recovery after small cortical ischemia. Stroke. 2004;35:992–7. doi: 10.1161/01.STR.0000119754.85848.0D. [DOI] [PubMed] [Google Scholar]
  • 272.Mizuno M, Yamada K, Olariu A, et al. Involvement of brain-derived neurotrophic factor in spatial memory formation and maintenance in a radial arm maze test in rats. J Neurosci. 2000;20:7116–21. doi: 10.1523/JNEUROSCI.20-18-07116.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Pencea V, Bingaman KD, Wiegand SJ, et al. Infusion of brain-derived neurotrophic factor into the lateral ventricle of the adult rat leads to new neurons in the parenchyma of the striatum, septum, thalamus, and hypothalamus. J Neurosci. 2001;21:6706–17. doi: 10.1523/JNEUROSCI.21-17-06706.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274.Zigova T, Pencea V, Wiegand SJ, et al. Intraventricular administration of BDNF increases the number of newly generated neurons in the adult olfactory bulb. Mol Cell Neurosci. 1998;11:234–45. doi: 10.1006/mcne.1998.0684. [DOI] [PubMed] [Google Scholar]
  • 275.Benraiss A, Chmielnicki E, Lerner K, et al. Adenoviral brain-derived neurotrophic factor induces both neostriatal and olfactory neuronal recruitment from endogenous progenitor cells in the adult forebrain. J Neurosci. 2001;21:6718–31. doi: 10.1523/JNEUROSCI.21-17-06718.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 276.Endres M, Fan G, Hirt L, et al. Ischemic brain damage in mice after selectively modifying BDNF or NT4 gene expression. J Cereb Blood Flow Metab. 2000;20:139–44. doi: 10.1097/00004647-200001000-00018. [DOI] [PubMed] [Google Scholar]
  • 277.Larsson E, Mandel RJ, Klein RL, et al. Suppression of insult-induced neurogenesis in adult rat brain by brain-derived neurotrophic factor. Exp Neurol. 2002;177:1–8. doi: 10.1006/exnr.2002.7992. [DOI] [PubMed] [Google Scholar]
  • 278.Andsberg G, Kokaia Z, Klein RL, et al. Neuropathological and behavioral consequences of adeno-associated viral vector-mediated continuous intrastriatal neurotrophin delivery in a focal ischemia model in rats. Neurobiol Dis. 2002;9:187–204. doi: 10.1006/nbdi.2001.0456. [DOI] [PubMed] [Google Scholar]
  • 279.Ferrer I, Krupinski J, Goutan E, et al. Brain-derived neurotrophic factor reduces cortical cell death by ischemia after middle cerebral artery occlusion in the rat. Acta Neuropathol. 2001;101:229–38. doi: 10.1007/s004010000268. [DOI] [PubMed] [Google Scholar]
  • 280.Schäbitz WR, Schwab S, Spranger M, et al. Intraventricular brain-derived neurotrophic factor reduces infarct size after focal cerebral ischemia in rats. J Cereb Blood Flow Metab. 1997;17:500–6. doi: 10.1097/00004647-199705000-00003. [DOI] [PubMed] [Google Scholar]
  • 281.Larsson E, Nanobashvili A, Kokaia Z, et al. Evidence for neuroprotective effects of endogenous brain-derived neurotrophic factor after global forebrain ischemia in rats. J Cereb Blood Flow Metab. 1999;19:1220–8. doi: 10.1097/00004647-199911000-00006. [DOI] [PubMed] [Google Scholar]
  • 282.Nygren J, Kokaia M, Wieloch T. Decreased expression of brain-derived neurotrophic factor in BDNF(+/−) mice is associated with enhanced recovery of motor performance and increased neuroblast number following experimental stroke. J Neurosci Res. 2006;84:626–31. doi: 10.1002/jnr.20956. [DOI] [PubMed] [Google Scholar]
  • 283.Pardridge WM, Wu D, Sakane T. Combined use of carboxyl-directed protein pegylation and vector-mediated blood-brain barrier drug delivery system optimizes brain uptake of brain-derived neurotrophic factor following intravenous administration. Pharm Res. 1998;15:576–82. doi: 10.1023/a:1011981927620. [DOI] [PubMed] [Google Scholar]
  • 284.Zhang Y, Pardridge WM. Blood-brain barrier targeting of BDNF improves motor function in rats with middle cerebral artery occlusion. Brain Res. 2006;1111:227–9. doi: 10.1016/j.brainres.2006.07.005. [DOI] [PubMed] [Google Scholar]
  • 285.Lin TN, Te J, Lee M, et al. Induction of basic fibroblast growth factor (bFGF) expression following focal cerebral ischemia. Brain Res Mol Brain Res. 1997;49:255–65. doi: 10.1016/s0169-328x(97)00152-6. [DOI] [PubMed] [Google Scholar]
  • 286.Zheng W, Nowakowski RS, Vaccarino FM. Fibroblast growth factor 2 is required for maintaining the neural stem cell pool in the mouse brain subventricular zone. Dev Neurosci. 2004;26:181–96. doi: 10.1159/000082136. [DOI] [PubMed] [Google Scholar]
  • 287.Cuevas P, Gonzalez AM, Carceller F, et al. Vascular response to basic fibroblast growth factor when infused onto the normal adventitia or into the injured media of the rat carotid artery. Circ Res. 1991;69:360–9. doi: 10.1161/01.res.69.2.360. [DOI] [PubMed] [Google Scholar]
  • 288.Rosenblatt S, Irikura K, Caday CG, et al. Basic fibroblast growth factor dilates rat pial arterioles. J Cereb Blood Flow Metab. 1994;14:70–4. doi: 10.1038/jcbfm.1994.11. [DOI] [PubMed] [Google Scholar]
  • 289.Naylor M, Bowen KK, Sailor KA, et al. Preconditioning-induced ischemic tolerance stimulates growth factor expression and neurogenesis in adult rat hippocampus. Neurochem Int. 2005;47:565–72. doi: 10.1016/j.neuint.2005.07.003. [DOI] [PubMed] [Google Scholar]
  • 290.Türeyen K, Vemuganti R, Bowen KK, et al. EGF and FGF-2 infusion increases post-ischemic neural progenitor cell proliferation in the adult rat brain. Neurosurgery. 2005;57:1254–63. doi: 10.1227/01.neu.0000186040.96929.8a. [DOI] [PubMed] [Google Scholar]
  • 291.Ay H, Ay I, Koroshetz WJ, et al. Potential usefulness of basic fibroblast growth factor as a treatment for stroke. Cerebrovasc Dis. 1999;9:131–5. doi: 10.1159/000015941. [DOI] [PubMed] [Google Scholar]
  • 292.Fisher M, Meadows ME, Do T, et al. Delayed treatment with intravenous basic fibroblast growth factor reduces infarct size following permanent focal cerebral ischemia in rats. J Cereb Blood Flow Metab. 1995;15:953–9. doi: 10.1038/jcbfm.1995.121. [DOI] [PubMed] [Google Scholar]
  • 293.Li Q, Stephenson D. Postischemic administration of basic fibroblast growth factor improves sensorimotor function and reduces infarct size following permanent focal cerebral ischemia in the rat. Exp Neurol. 2002;177:531–7. doi: 10.1006/exnr.2002.7994. [DOI] [PubMed] [Google Scholar]
  • 294.Wada K, Sugimori H, Bhide PG, et al. Effect of basic fibroblast growth factor treatment on brain progenitor cells after permanent focal ischemia in rats. Stroke. 2003;34:2722–8. doi: 10.1161/01.STR.0000094421.61917.71. [DOI] [PubMed] [Google Scholar]
  • 295.Türeyen K, Vemuganti R, Bowen KK, et al. EGF and FGF-2 infusion increases post-ischemic neural progenitor cell proliferation in the adult rat brain. Neurosurgery. 2005;57:1254–63. doi: 10.1227/01.neu.0000186040.96929.8a. [DOI] [PubMed] [Google Scholar]
  • 296.Baldauf K, Reymann KG. Influence of EGF/bFGF treatment on proliferation, early neurogenesis and infarct volume after transient focal ischemia. Brain Res. 2005;1056:158–67. doi: 10.1016/j.brainres.2005.07.035. [DOI] [PubMed] [Google Scholar]
  • 297.Ikeda N, Nonoguchi N, Zhao MZ, et al. Bone marrow stromal cells that enhanced fibroblast growth factor-2 secretion by herpes simplex virus vector improve neurological outcome after transient focal cerebral ischemia in rats. Stroke. 2005;36:2725–30. doi: 10.1161/01.STR.0000190006.88896.d3. [DOI] [PubMed] [Google Scholar]
  • 298.Watanabe T, Okuda Y, Nonoguchi N, et al. Postischemic intraventricular administration of FGF-2 expressing adenoviral vectors improves neurologic outcome and reduces infarct volume after transient focal cerebral ischemia in rats. J Cereb Blood Flow Metab. 2004;24:1205–13. doi: 10.1097/01.WCB.0000136525.75839.41. [DOI] [PubMed] [Google Scholar]
  • 299.Wahlgren NG, Ahmed N. Neuroprotection in cerebral ischaemia: facts and fancies – the need for new approaches. Cerebrovasc Dis. 2004;17:153–66. doi: 10.1159/000074808. [DOI] [PubMed] [Google Scholar]
  • 300.Bogousslavsky J, Victor SJ, Salinas EO, et al. European-Australian Fiblast (Trafermin) in Acute Stroke Group Fiblast (trafermin) in acute stroke: results of the European-Australian phase II/III safety and efficacy trial. Cerebrovasc Dis. 2002;14:239–51. doi: 10.1159/000065683. [DOI] [PubMed] [Google Scholar]
  • 301.Guo H, Huang L, Cheng M, et al. Serial measurement of serum basic fibroblast growth factor in patients with acute cerebral infarction. Neurosci Lett. 2006;393:56–9. doi: 10.1016/j.neulet.2005.09.043. [DOI] [PubMed] [Google Scholar]
  • 302.Endres M, Piriz J, Gertz K, et al. Serum insulin-like growth factor I and ischemic brain injury. Brain Res. 2007;1185:328–35. doi: 10.1016/j.brainres.2007.09.053. [DOI] [PubMed] [Google Scholar]
  • 303.Pardridge WM. Transport of insulin-related peptides and glucose across the blood–brain barrier. Ann NY Acad Sci. 1993;692:126–37. doi: 10.1111/j.1749-6632.1993.tb26211.x. [DOI] [PubMed] [Google Scholar]
  • 304.Reinhardt RR, Bondy CA. Insulin-like growth factors cross the blood–brain barrier. Endocrinology. 1994;135:1753–61. doi: 10.1210/endo.135.5.7525251. [DOI] [PubMed] [Google Scholar]
  • 305.Armstrong CS, Wuarin L, Ishii DN. Uptake of circulating insulin-like growth factor-I into the cerebrospinal fluid of normal and diabetic rats and normalization of IGF-II mRNA content in diabetic rat brain. J Neurosci Res. 2000;59:649–60. doi: 10.1002/(SICI)1097-4547(20000301)59:5<649::AID-JNR8>3.0.CO;2-W. [DOI] [PubMed] [Google Scholar]
  • 306.Carro E, Nunez A, Busiguina S, et al. Circulating insulin-like growth factor I mediates effects of exercise on the brain. J Neurosci. 2000;20:2926–33. doi: 10.1523/JNEUROSCI.20-08-02926.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 307.Torres-Aleman I. Serum growth factors and neuroprotective surveillance. Mol Neurobiol. 2000;21:153–60. doi: 10.1385/mn:21:3:153. [DOI] [PubMed] [Google Scholar]
  • 308.Nunez A, Carro E, Torres-Aleman I. Insulin-like growth factor I modifies electrophysiological properties of rat brain stem neurons. J. Neurophysiol. 2003;89:3008–17. doi: 10.1152/jn.00089.2003. [DOI] [PubMed] [Google Scholar]
  • 309.Dunn SE. Insulin-like growth factor I stimulates angiogenesis and the production of vascular endothelial growth factor. Growth Horm IGF Res. 2000;10:S41–2. doi: 10.1016/s1096-6374(00)90020-0. [DOI] [PubMed] [Google Scholar]
  • 310.Leinninger GM, Feldman EL. Insulin-like growth factors in the treatment of neurological disease. Endocr Dev. 2005;9:135–59. doi: 10.1159/000085763. [DOI] [PubMed] [Google Scholar]
  • 311.Hellström A, Carlsson B, Niklasson A, et al. IGF-I is critical for normal vascularization of the human retina. J Clin Endocrinol Metab. 2002;87:3413–6. doi: 10.1210/jcem.87.7.8629. [DOI] [PubMed] [Google Scholar]
  • 312.Sonntag WE, Lynch CD, Cooney PT, et al. Decreases in cerebral microvasculature with age are associated with the decline in growth hormone and insulin-like growth factor 1. Endocrinology. 1997;138:3515–20. doi: 10.1210/endo.138.8.5330. [DOI] [PubMed] [Google Scholar]
  • 313.Conti E, Carrozza C, Capoluongo E, et al. Insulin-like growth factor-1 as a vascular protective factor. Circulation. 2004;110:2260–5. doi: 10.1161/01.CIR.0000144309.87183.FB. [DOI] [PubMed] [Google Scholar]
  • 314.Schwab S, Spranger M, Krempien S, et al. Plasma insulin-like growth factor I and IGF binding protein 3 levels in patients with acute cerebral ischemic injury. Stroke. 1997;28:1744–8. doi: 10.1161/01.str.28.9.1744. [DOI] [PubMed] [Google Scholar]
  • 315.Denti L, Annoni V, Cattadori E, et al. Insulin-like growth factor 1 as a predictor of ischemic stroke outcome in the elderly. Am J Med. 2004;117:312–7. doi: 10.1016/j.amjmed.2004.02.049. [DOI] [PubMed] [Google Scholar]
  • 316.Strasburger CJ, Bidlingmaier M, Wu Z, et al. Normal values of insulin-like growth factor I and their clinical utility in adults. Horm Res. 2001;55:100–5. doi: 10.1159/000063484. [DOI] [PubMed] [Google Scholar]
  • 317.Beilharz EJ, Russo VC, Butler G, et al. Co-ordinated and cellular specific induction of the components of the IGF/IGFBP axis in the rat brain following hypoxic-ischemic injury. Brain Res Mol Brain Res. 1998;59:119–34. doi: 10.1016/s0169-328x(98)00122-3. [DOI] [PubMed] [Google Scholar]
  • 318.Guan J, Williams CE, Skinner SJ, et al. The effects of insulin-like growth factor (IGF)-1, IGF-2, and des-IGF-1 on neuronal loss after hypoxic-ischemic brain injury in adult rats: evidence for a role for IGF binding proteins. Endocrinology. 1996;137:893–8. doi: 10.1210/endo.137.3.8603600. [DOI] [PubMed] [Google Scholar]
  • 319.Johnston BM, Mallard EC, Williams CE, et al. Insulin-like growth factor-1 is a potent neuronal rescue agent after hypoxic-ischemic injury in fetal lambs. J Clin Invest. 1996;97:300–8. doi: 10.1172/JCI118416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 320.Schabitz WR, Hoffmann TT, Heiland S, et al. Delayed neuroprotective effect of insulin-like growth factor-I after experimental transient focal cerebral ischemia monitored with MRI. Stroke. 2001;32:1226–33. doi: 10.1161/01.str.32.5.1226. [DOI] [PubMed] [Google Scholar]
  • 321.Liu XF, Fawcett JR, Thorne RG, et al. Intranasal administration of insulin-like growth factor-I bypasses the blood– brain barrier and protects against focal cerebral ischemic damage. J Neurol Sci. 2001;187:91–7. doi: 10.1016/s0022-510x(01)00532-9. [DOI] [PubMed] [Google Scholar]
  • 322.Zhu W, Fan Y, Frenzel T, et al. Insulin growth factor-1 gene transfer enhances neurovascular remodeling and improves long-term stroke outcome in mice. Stroke. 2008;39:1254–61. doi: 10.1161/STROKEAHA.107.500801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 323.Rizk NN, Myatt-Jones J, Rafols J, et al. Insulin like growth factor-1 (IGF-1) decreases ischemia-reperfusion induced apoptosis and necrosis in diabetic rats. Endocrine. 2007;31:66–71. doi: 10.1007/s12020-007-0012-0. [DOI] [PubMed] [Google Scholar]
  • 324.Lin LF, Doherty DH, Lile JD, et al. GDNF: a glial cell line-derived neurotrophic factor for midbrain dopaminergic neurones. Science. 1993;260:1130–2. doi: 10.1126/science.8493557. [DOI] [PubMed] [Google Scholar]
  • 325.Ramer MS, Priestley JV, McMahon SB. Functional regeneration of sensory axons into the adult spinal cord. Nature. 2000;403:312–6. doi: 10.1038/35002084. [DOI] [PubMed] [Google Scholar]
  • 326.Henderson CE, Phillips HS, Pollock RA, et al. GDNF: a potent survival factor for motoneurons present in peripheral nerve and muscle. Science. 1994;266:1062–4. doi: 10.1126/science.7973664. [DOI] [PubMed] [Google Scholar]
  • 327.Arenas E, Trupp M, Akerud P, et al. GDNF prevents degeneration and promotes the phenotype of brain noradrenergicneurons in vivo. Neuron. 1995;15:1465–73. doi: 10.1016/0896-6273(95)90024-1. [DOI] [PubMed] [Google Scholar]
  • 328.Airaksinen MS, Saarma M. The GDNF family: signalling, biological functions and therapeutic value. Nat Rev Neurosci. 2002;3:383–94. doi: 10.1038/nrn812. [DOI] [PubMed] [Google Scholar]
  • 329.Kitagawa H, Hayashi T, Mitsumoto Y, et al. Reduction of ischemic brain injury by topical application of glial cell line-derived neurotrophic factor after permanent middle cerebral artery occlusion in rats. Stroke. 1998;29:1417–22. doi: 10.1161/01.str.29.7.1417. [DOI] [PubMed] [Google Scholar]
  • 330.Wang Y, Lin SZ, Chiou AL, et al. Glial cell line derived neurotrophic factor protects against ischemia-induced injury in the cerebral cortex. J Neurosci. 1997;17:4341–8. doi: 10.1523/JNEUROSCI.17-11-04341.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 331.Zhang WR, Sato K, Iwai M, et al. Therapeutic time window of adenovirus-mediated GDNF gene transfer after transient middle cerebral artery occlusion in rat. Brain Res. 2002;947:140–5. doi: 10.1016/s0006-8993(02)02923-2. [DOI] [PubMed] [Google Scholar]
  • 332.Arvidsson A, Kokaia Z, Airaksinen MS, et al. Stroke induces widespread changes of gene expression for glial cell line-derived neurotrophic factor family receptors in the adult rat brain. Neurosci. 2001;106:27–41. doi: 10.1016/s0306-4522(01)00268-8. [DOI] [PubMed] [Google Scholar]
  • 333.Arvidsson A, Kirik D, Lundberg C, et al. Elevated GDNF levels following viral vector-mediated gene transfer can increase neuronal death after stroke in rats. Neurobiol Dis. 2003;14:542–56. doi: 10.1016/j.nbd.2003.08.002. [DOI] [PubMed] [Google Scholar]
  • 334.Dempsey RJ, Sailor KA, Bowen KK, et al. Stroke-induced progenitor cell proliferation in adult spontaneously hypertensive rat brain: effect of exogenous IGF-1 and GDNF. J Neurochem. 2003;87:586–97. doi: 10.1046/j.1471-4159.2003.02022.x. [DOI] [PubMed] [Google Scholar]
  • 335.Kobayashi T, Ahlenius H, Thored P, et al. Intracerebral infusion of glial cell line-derived neurotrophic factor promotes striatal neurogenesis after stroke in adult rats. Stroke. 2006;37:2361–7. doi: 10.1161/01.STR.0000236025.44089.e1. [DOI] [PubMed] [Google Scholar]
  • 336.Horita Y, Honmou O, Harada K, et al. Intravenous administration of glial cell line-derived neurotrophic factor gene-modified human mesenchymal stem cells protects against injury in a cerebral ischemia model in the adult rat. J Neurosci Res. 2006;84:1495–504. doi: 10.1002/jnr.21056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 337.Higashiyama S, Abraham JA, Miller J, et al. A heparin-binding growth factor secreted by macrophage-like cells that is related to EGF. Science. 1991;251:936–9. doi: 10.1126/science.1840698. [DOI] [PubMed] [Google Scholar]
  • 338.Goishi K, Higashiyama S, Klagsbrun M, et al. Phorbol ester induces the rapid processing of cell surface heparin-binding EGF-like growth factor: conversion from juxtacrine to paracrine growth factor activity. Mol Biol Cell. 1995;6:967–80. doi: 10.1091/mbc.6.8.967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 339.Mishima K, Higashiyama S, Nagashima Y, et al. Regional distribution of heparin-binding epidermal growth factor-like growth factor mRNA and protein in adult rat forebrain. Neurosci Lett. 1996;213:153–6. doi: 10.1016/0304-3940(96)12850-0. [DOI] [PubMed] [Google Scholar]
  • 340.Morrison RS, Kornblum HI, Leslie FM, et al. Trophic stimulation of cultured neurons from neonatal rat brain by epidermal growth factor. Science. 1987;238:72–5. doi: 10.1126/science.3498986. [DOI] [PubMed] [Google Scholar]
  • 341.Maiese K, Boniece I, DeMeo D, et al. Peptide growth factors protect against ischemia in culture by preventing nitric oxide toxicity. J Neurosci. 1993;13:3034–40. doi: 10.1523/JNEUROSCI.13-07-03034.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 342.Kawahara N, Mishima K, Higashiyama S, et al. The gene for heparin-binding epidermal growth factor-like growth factor is stress-inducible: its role in cerebral ischemia. J Cereb Blood Flow Metab. 1999;19:307–20. doi: 10.1097/00004647-199903000-00009. [DOI] [PubMed] [Google Scholar]
  • 343.Teramoto T, Qiu J, Plumier JC, et al. EGF amplifies the replacement of parvalbumin-expressing striatal interneurons after ischemia. J Clin Invest. 2003;111:1125–32. doi: 10.1172/JCI17170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 344.Nakatomi H, Kuriu T, Okabe S, et al. Regeneration of hippocampal pyramidal neurons after ischemic brain injury by recruitment of endogenous neural progenitors. Cell. 2002;110:429–41. doi: 10.1016/s0092-8674(02)00862-0. [DOI] [PubMed] [Google Scholar]
  • 345.Sugiura S, Kitagawa K, Tanaka S, et al. Adenovirus-mediated gene transfer of heparin-binding epidermal growth factorlike growth factor enhances neurogenesis and angiogenesis after focal cerebral ischemia in rats. Stroke. 2005;36:859–64. doi: 10.1161/01.STR.0000158905.22871.95. [DOI] [PubMed] [Google Scholar]
  • 346.Zhang ZG, Chopp M. Neurorestorative therapies for stroke: underlying mechanisms and translation to the clinic. Lancet Neurol. 2009;8:491–500. doi: 10.1016/S1474-4422(09)70061-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 347.Jin K, Mao XO, Sun Y, et al. Heparin-binding epidermal growth factor-like growth factor: hypoxiainducible expression in vitro and stimulation of neurogenesis in vitro and in vivo. J Neurosci. 2002;22:5365–73. doi: 10.1523/JNEUROSCI.22-13-05365.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 348.Reuss B, von Bohlen, Halbach O. Fibroblast growth factors and their receptors in the central nervous system. Cell Tissue Res. 2003;313:139–57. doi: 10.1007/s00441-003-0756-7. [DOI] [PubMed] [Google Scholar]
  • 349.Takami K, Kiyota Y, Iwane M, et al. Upregulation of fibroblast growth factor-receptor messenger RNA expression in rat brain following transient forebrain ischemia. Exp Brain Res. 1993;97:185–94. doi: 10.1007/BF00228688. [DOI] [PubMed] [Google Scholar]
  • 350.Wong RW, Guillaud L. The role of epidermal growth factor and its receptors in mammalian CNS. Cytokine Growth Factor Rev. 2004;15:147–56. doi: 10.1016/j.cytogfr.2004.01.004. [DOI] [PubMed] [Google Scholar]
  • 351.Hanson LR, Frey WH. Intranasal delivery bypasses the blood-brain barrier to target therapeutic agents to the central nervous system and treat neurodegenerative disease. BMC Neurosci. 2008;9:S5. doi: 10.1186/1471-2202-9-S3-S5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 352.Piao CS, Gonzalez-Toledo ME, Xue YQ, et al. The role of stem cell factor and granulocyte-colony stimulating factor in brain repair during chronic stroke. J Cereb Blood Flow Metab. 2009;29:759–70. doi: 10.1038/jcbfm.2008.168. [DOI] [PubMed] [Google Scholar]

Articles from Journal of Cellular and Molecular Medicine are provided here courtesy of Blackwell Publishing

RESOURCES